40 results on '"Ronald J. Holewinski"'
Search Results
2. Abstracts from the 8th International Conference on cGMP Generators, Effectors and Therapeutic Implications
- Author
-
G. Todd Milne, on behalf of the Ironwood team, Peter Sandner, Kathleen A. Lincoln, Paul C. Harrison, Hongxing Chen, Hong Wang, Holly Clifford, Hu Sheng Qian, Diane Wong, Chris Sarko, Ryan Fryer, Jeremy Richman, Glenn A. Reinhart, Carine M. Boustany, Steven S. Pullen, Henriette Andresen, Lise Román Moltzau, Alessandro Cataliotti, Finn Olav Levy, Robert Lukowski, Sandra Frankenreiter, Andreas Friebe, Timothy Calamaras, Robert Baumgartner, Angela McLaughlin, Mark Aronovitz, Wendy Baur, Guang-Rong Wang, Navin Kapur, Richard Karas, Robert Blanton, Stefan Hell, Scott A. Waldman, Jieru E. Lin, Francheska Colon-Gonzalez, Gilbert W. Kim, Erik S. Blomain, Dante Merlino, Adam Snook, Jeanette Erdmann, Jana Wobst, Thorsten Kessler, Heribert Schunkert, Ulrich Walter, Oliver Pagel, Elena Walter, Stepan Gambaryan, Albert Smolenski, Kerstin Jurk, Rene Zahedi, James R. Klinger, Raymond L. Benza, Paul A. Corris, David Langleben, Robert Naeije, Gérald Simonneau, Christian Meier, Pablo Colorado, Mi Kyung Chang, Dennis Busse, Marius M. Hoeper, Jaime L. Masferrer, Sarah Jacobson, Guang Liu, Renee Sarno, Sylvie Bernier, Ping Zhang, Roger Flores-Costa, Mark Currie, Katherine Hall, Dorit Möhrle, Katrin Reimann, Steffen Wolter, Markus Wolters, Evanthia Mergia, Nicole Eichert, Hyun-Soon Geisler, Peter Ruth, Robert Feil, Ulrike Zimmermann, Doris Koesling, Marlies Knipper, Lukas Rüttiger, Yasutake Tanaka, Atsuko Okamoto, Takashi Nojiri, Motofumi Kumazoe, Takeshi Tokudome, Koichi Miura, Jun Hino, Hiroshi Hosoda, Mikiya Miyazato, Kenji Kangawa, Vikas Kapil, Amrita Ahluwalia, Nazareno Paolocci, Philip Eaton, James C. Campbell, Philipp Henning, Eugen Franz, Banumathi Sankaran, Friedrich W. Herberg, Choel Kim, M. Wittwer, Q. Luo, V. Kaila, S. A. Dames, Andrew Tobin, Mahmood Alam, Olena Rudyk, Susanne Krasemann, Kristin Hartmann, Oleksandra Prysyazhna, Min Zhang, Lan Zhao, Astrid Weiss, Ralph Schermuly, Amie J. Moyes, Sandy M. Chu, Reshma S. Baliga, Adrian J. Hobbs, Stylianos Michalakis, Regine Mühlfriedel, Christian Schön, Dominik M. Fischer, Barbara Wilhelm, Ditta Zobor, Susanne Kohl, Tobias Peters, Eberhart Zrenner, Karl Ulrich Bartz-Schmidt, Marius Ueffing, Bernd Wissinger, Mathias Seeliger, Martin Biel, RD-CURE consortium, Mark J. Ranek, Kristen M. Kokkonen, Dong I. Lee, Ronald J. Holewinski, Vineet Agrawal, Cornelia Virus, Donté A. Stevens, Masayuki Sasaki, Huaqun Zhang, Mathew M. Mannion, Peter P. Rainer, Richard C. Page, Jonathan C. Schisler, Jennifer E. Van Eyk, Monte S. Willis, David A. Kass, Manuela Zaccolo, Michael Russwurm, Jan Giesen, Corina Russwurm, Ernst-Martin Füchtbauer, Nadja I. Bork, Viacheslav O. Nikolaev, Luis Agulló, Martin Floor, Jordi Villà-Freixa, Ornella Manfra, Gaia Calamera, Nicoletta C. Surdo, Silja Meier, Alexander Froese, Kjetil Wessel Andressen, Annemarie Aue, Fabian Schwiering, Dieter Groneberg, Gzona Bajraktari, Jürgen Burhenne, Walter E. Haefeli, Johanna Weiss, Katharina Beck, Barbara Voussen, Alexander Vincent, Sean P. Parsons, Jan D. Huizinga, Fabiola Zakia Mónica, Edward Seto, Ferid Murad, Ka Bian, Joseph R. Burgoyne, Daniel Richards, Marianne Bjørnerem, Andrea Hembre Ulsund, Jeong Joo Kim, Sonia Donzelli, Mara Goetz, Kjestine Schmidt, Konstantina Stathopoulou, Jenna Scotcher, Christian Dees, Hariharan Subramanian, Elke Butt, Alisa Kamynina, S. Bruce King, Cor de Witt, Lars I. Leichert, Friederike Cuello, Hyazinth Dobrowinski, Moritz Lehners, Michael Paolillo Hannes Schmidt, Susanne Feil, Lai Wen, Martin Thunemann, Marcus Olbrich, Harald Langer, Meinrad Gawaz, Cor de Wit, Daniela Bertinetti, Hossein-Ardeschir Ghofrani, Friedrich Grimminger, Ekkehard Grünig, Yigao Huang, Pavel Jansa, Zhi Cheng Jing, David Kilpatrick, Stephan Rosenkranz, Flavia Menezes, Arno Fritsch, Sylvia Nikkho, Reiner Frey, Marc Humbert, Manuela Harloff, Joerg Reinders, Jens Schlossmann, Joon Jung, Jessica A. Wales, Cheng-Yu Chen, Linda Breci, Andrzej Weichsel, Sylvie G. Bernier, Robert Solinga, James E. Sheppeck, Paul A. Renhowe, William R. Montfort, Liying Qin, Ying-Ju Sung, Darren Casteel, Alexander Kollau, Andrea Neubauer, Astrid Schrammel, Bernd Mayer, Mika Takai, Chieri Takeuchi, Mai Kadomatsu, Shun Hiroi, Kanako Takamatsu, Hirofumi Tachibana, Marissa Opelt, Emrah Eroglu, Markus Waldeck-Weiermair, Roland Malli, Wolfgang F. Graier, John T. Fassett, Selene J. Sollie, Maria Hernandez-Valladares, Frode Berven, Kjetil W. Andressen, Miki Arai, Yutaka Suzuki, Meinoshin Okumura, Shinpei Kawaoka, Stefanie Peters, Hannes Schmidt, B. Selin Kenet, Sarah Helena Nies, Katharina Frank, Fritz G. Rathjen, Olga N. Petrova, Isabelle Lamarre, Michel Négrerie, Jerid W. Robinson, Jeremy R. Egbert, Julia Davydova, Laurinda A. Jaffe, Lincoln R. Potter, Nicholas Blixt, Leia C. Shuhaibar, Gordon L. Warren, Kim C. Mansky, Simone Romoli, Tobias Bauch, Karoline Dröbner, Frank Eitner, Mihály Ruppert, Tamás Radovits, Sevil Korkmaz-Icöz, Shiliang Li, Péter Hegedűs, Sivakanan Loganathan, Balázs Tamás Németh, Attila Oláh, Csaba Mátyás, Kálmán Benke, Béla Merkely, Matthias Karck, Gábor Szabó, Ulrike Scheib, Matthias Broser, Shatanik Mukherjee, Katja Stehfest, Christine E. Gee, Heinz G. Körschen, Thomas G. Oertner, Peter Hegemann, Deborah M. Dickey, Alexandre Dumoulin, Ralf Kühn, Laurinda Jaffe, Sophie Schobesberger, Peter Wright, Claire Poulet, Catherine Mansfield, Sian E. Harding, Julia Gorelik, Gerald Wölkart, Antonius C. F. Gorren, Gerburg K. Schwaerzer, Darren E. Casteel, Nancy D. Dalton, Yusu Gu, Shunhui Zhuang, Dianna M. Milewicz, Kirk L. Peterson, Renate Pilz, Aikaterini I. Argyriou, Garyfalia Makrynitsa, Ioannis I. Alexandropoulos, Andriana Stamopoulou, Marina Bantzi, Athanassios Giannis, Stavros Topouzis, Andreas Papapetropoulos, Georgios A. Spyroulias, Dennis J. Stuehr, Arnab Ghosh, Yue Dai, Saurav Misra, Boris Tchernychev, Inmaculada Silos-Santiago, Gerhard Hannig, Vu Thao-Vi Dao, Martin Deile, Pavel I. Nedvetsky, Andreas Güldner, César Ibarra-Alvarado, Axel Gödecke, Harald H. H. W. Schmidt, Angelos Vachaviolos, Andrea Gerling, Stefan Z. Lutz, Hans-Ulrich Häring, Marcel A. Krüger, Bernd J. Pichler, Michael J. Shipston, Sara Vandenwijngaert, Clara D. Ledsky, Obiajulu Agha, Dongjian Hu, Ibrahim J. Domian, Emmanuel S. Buys, Christopher Newton-Cheh, Donald B. Bloch, Nadine Mauro, Jonas Keppler, Wilson A. Ferreira, Hanan Chweih, Pamela L. Brito, Camila B. Almeida, Carla F. F. Penteado, Sara S. O. Saad, Fernando F. Costa, Paul S. Frenette, Damian Brockschnieder, Johannes-Peter Stasch, Nicola Conran, Daniel P. Zimmer, Jenny Tobin, Courtney Shea, Kimberly Long, Kim Tang, Peter Germano, James Wakefield, Ali Banijamali, G-Yoon Jamie Im, Albert T. Profy, and Mark G. Currie
- Subjects
Therapeutics. Pharmacology ,RM1-950 ,Toxicology. Poisons ,RA1190-1270 - Published
- 2017
- Full Text
- View/download PDF
3. Gene and protein expression in human megakaryocytes derived from induced pluripotent stem cells
- Author
-
Nauder Faraday, Kai Kammers, Linzhao Cheng, Lisa R. Yanek, Joshua Martin, Diane M. Becker, Jennifer E. Van Eyk, Senquan Liu, Victoria Dardov, Zack Z. Wang, Lewis C. Becker, Jeffrey T. Leek, Margaret A. Taub, Ronald J. Holewinski, Niveda Sundararaman, Koen Raedschelders, Rasika A. Mathias, Kanika Kanchan, Vidya Venkatraman, Dixie L. Hoyle, and Sarah J. Parker
- Subjects
Messenger RNA ,urogenital system ,CD34 ,Hematology ,030204 cardiovascular system & hematology ,Biology ,Peripheral blood mononuclear cell ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,Gene expression ,Progenitor cell ,Induced pluripotent stem cell ,Gene - Abstract
Background There is interest in deriving megakaryocytes (MKs) from pluripotent stem cells (iPSC) for biological studies. We previously found that genomic structural integrity and genotype concordance is maintained in iPSC-derived MKs. Objective To establish a comprehensive dataset of genes and proteins expressed in iPSC-derived MKs. Methods iPSCs were reprogrammed from peripheral blood mononuclear cells (MNCs) and MKs were derived from the iPSCs in 194 healthy European American and African American subjects. mRNA was isolated and gene expression measured by RNA sequencing. Protein expression was measured in 62 of the subjects using mass spectrometry. Results and conclusions MKs expressed genes and proteins known to be important in MK and platelet function and demonstrated good agreement with previous studies in human MKs derived from CD34+ progenitor cells. The percent of cells expressing the MK markers CD41 and CD42a was consistent in biological replicates, but variable across subjects, suggesting that unidentified subject-specific factors determine differentiation of MKs from iPSCs. Gene and protein sets important in platelet function were associated with increasing expression of CD41/42a, while those related to more basic cellular functions were associated with lower CD41/42a expression. There was differential gene expression by the sex and race (but not age) of the subject. Numerous genes and proteins were highly expressed in MKs but not known to play a role in MK or platelet function; these represent excellent candidates for future study of hematopoiesis, platelet formation, and/or platelet function.
- Published
- 2021
4. Lysine and Arginine Protein Post-translational Modifications by Enhanced DIA Libraries: Quantification in Murine Liver Disease
- Author
-
Shelly C. Lu, Vidya Venkatraman, George Rosenberger, Gautam Saxena, Aleksandra Binek, Brian C. Searle, Nathan Basisty, Birgit Schilling, Ronald J. Holewinski, Jennifer E. Van Eyk, Benjamin A. Garcia, Peder J. Lund, Aaron E. Robinson, Xueshu Xie, Sarah J. Parker, and José M. Mato
- Subjects
Proteomics ,0301 basic medicine ,chemistry.chemical_classification ,Messenger RNA ,030102 biochemistry & molecular biology ,Arginine ,Liver Diseases ,Lysine ,Acetylation ,Peptide ,General Chemistry ,Methylation ,Biochemistry ,Mice ,03 medical and health sciences ,Succinylation ,030104 developmental biology ,chemistry ,Protein biosynthesis ,Animals ,Protein Processing, Post-Translational - Abstract
Proteoforms containing post-translational modifications (PTMs) represent a degree of functional diversity only harnessed through analytically precise simultaneous quantification of multiple PTMs. Here we present a method to accurately differentiate an unmodified peptide from its PTM-containing counterpart through data-independent acquisition-mass spectrometry, leveraging small precursor mass windows to physically separate modified peptidoforms from each other during MS2 acquisition. We utilize a lysine and arginine PTM-enriched peptide assay library and site localization algorithm to simultaneously localize and quantify seven PTMs including mono-, di-, and trimethylation, acetylation, and succinylation in addition to total protein quantification in a single MS run without the need to enrich experimental samples. To evaluate biological relevance, this method was applied to liver lysate from differentially methylated nonalcoholic steatohepatitis (NASH) mouse models. We report that altered methylation and acetylation together with total protein changes drive the novel hypothesis of a regulatory function of PTMs in protein synthesis and mRNA stability in NASH.
- Published
- 2020
5. Comparative Proteomic Analysis of HPV(+) Oropharyngeal Squamous Cell Carcinoma Recurrence
- Author
-
Stephen L. Shiao, Koen Raedschelders, Jon Mallen-St. Clair, Yi Zhang, Wonwoo Shon, Vidya Venkatraman, Jennifer E. Van Eyk, Anna Laury, Aaron E. Robinson, Michelle M. Chen, Allen S. Ho, Ronald J. Holewinski, Zachary S. Zumsteg, and De-Chen Lin
- Subjects
Proteomics ,Programmed cell death ,RAD23B ,Nerve Tissue Proteins ,Biochemistry ,Profilins ,Medicine ,Humans ,Papillomaviridae ,business.industry ,Squamous Cell Carcinoma of Head and Neck ,Head and neck cancer ,Papillomavirus Infections ,Wnt signaling pathway ,General Chemistry ,medicine.disease ,Prognosis ,Fold change ,Staining ,DNA-Binding Proteins ,Oropharyngeal Neoplasms ,DNA Repair Enzymes ,Apoptosis ,Head and Neck Neoplasms ,Cancer research ,Immunohistochemistry ,business - Abstract
Deintensification therapy for human papillomavirus-related oropharyngeal squamous cell carcinoma (HPV(+) OPSCC) is under active investigation. An adaptive treatment approach based on molecular stratification could identify high-risk patients predisposed to recurrence and better select for appropriate treatment regimens. Collectively, 40 HPV(+) OPSCC FFPE samples (20 disease-free, 20 recurrent) were surveyed using mass spectrometry-based proteomic analysis via data-independent acquisition to obtain fold change and false discovery differences. Ten-year overall survival was 100.0 and 27.7% for HPV(+) disease-free and recurrent cohorts, respectively. Of 1414 quantified proteins, 77 demonstrated significant differential expression. Top enriched functional pathways included those involved in programmed cell death (73 proteins, p = 7.43 × 10-30), apoptosis (73 proteins, p = 5.56 × 10-9), β-catenin independent WNT signaling (47 proteins, p = 1.45 × 10-15), and Rho GTPase signaling (69 proteins, p = 1.09 × 10-5). PFN1 (p = 1.0 × 10-3), RAD23B (p = 2.9 × 10-4), LDHB (p = 1.0 × 10-3), and HINT1 (p = 3.8 × 10-3) pathways were significantly downregulated in the recurrent cohort. On functional validation via immunohistochemistry (IHC) staining, 46.9% (PFN1), 71.9% (RAD23B), 59.4% (LDHB), and 84.4% (HINT1) of cases were corroborated with mass spectrometry findings. Development of a multilateral molecular signature incorporating these targets may characterize high-risk disease, predict treatment response, and augment current management paradigms in head and neck cancer.
- Published
- 2021
6. Myofilament Phosphorylation in Stem Cell Treated Diastolic Heart Failure
- Author
-
Daniel Soetkamp, Joshua I. Goldhaber, Jennifer E. Van Eyk, Sarah J. Parker, Eduardo Marbán, Ronald J. Holewinski, Kiel Peck, Romain Gallet, and Vidya Venkatraman
- Subjects
Male ,medicine.medical_specialty ,Myofilament ,Physiology ,Clinical Sciences ,Diastole ,Cardiorespiratory Medicine and Haematology ,Cardiovascular ,Article ,Cell Line ,Myofibrils ,Fibrosis ,Internal medicine ,medicine ,echocardiography ,Animals ,Phosphorylation ,Protein Kinase C ,mass spectrometry ,Heart Failure ,Rats, Inbred Dahl ,business.industry ,phosphorylation ,fibrosis ,Diastolic heart failure ,medicine.disease ,Rats ,Heart Disease ,Cardiovascular System & Hematology ,Heart failure ,Inbred Dahl ,Cardiology ,Stem cell ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business ,Stem Cell Transplantation - Abstract
Rationale: Phosphorylation of sarcomeric proteins has been implicated in heart failure with preserved ejection fraction (HFpEF); such changes may contribute to diastolic dysfunction by altering contractility, cardiac stiffness, Ca 2+ -sensitivity, and mechanosensing. Treatment with cardiosphere-derived cells (CDCs) restores normal diastolic function, attenuates fibrosis and inflammation, and improves survival in a rat HFpEF model. Objective: Phosphorylation changes that underlie HFpEF and those reversed by CDC therapy, with a focus on the sarcomeric subproteome were analyzed. Methods and Results: Dahl salt–sensitive rats fed a high-salt diet, with echocardiographically verified diastolic dysfunction, were randomly assigned to either intracoronary CDCs or placebo. Dahl salt–sensitive rats receiving low salt diet served as controls. Protein and phosphorylated Ser, Thr, and Tyr residues from left ventricular tissue were quantified by mass spectrometry. HFpEF hearts exhibited extensive hyperphosphorylation with 98% of the 529 significantly changed phospho-sites increased compared with control. Of those, 39% were located within the sarcomeric subproteome, with a large group of proteins located or associated with the Z-disk. CDC treatment partially reverted the hyperphosphorylation, with 85% of the significantly altered 76 residues hypophosphorylated. Bioinformatic upstream analysis of the differentially phosphorylated protein residues revealed PKC as the dominant putative regulatory kinase. PKC isoform analysis indicated increases in PKC α, β, and δ concentration, whereas CDC treatment led to a reversion of PKCβ. Use of PKC isoform specific inhibition and overexpression of various PKC isoforms strongly suggests that PKCβ is the dominant kinase involved in hyperphosphorylation in HFpEF and is altered with CDC treatment. Conclusions: Increased protein phosphorylation at the Z-disk is associated with diastolic dysfunction, with PKC isoforms driving most quantified phosphorylation changes. Because CDCs reverse the key abnormalities in HFpEF and selectively reverse PKCβ upregulation, PKCβ merits being classified as a potential therapeutic target in HFpEF, a disease notoriously refractory to medical intervention.
- Published
- 2021
7. Mechanistic dissection of global proteomic changes in rats with heart failure and preserved ejection fraction
- Author
-
Daniel Soetkamp, Peter J. Kilfoil, Aleksandra Binek, G. de Couto, J. E. Van Eyk, Amy D. Bradshaw, Joshua I. Goldhaber, Eduardo Marbán, Ronald J. Holewinski, Vidya Venkatraman, Rowann Mostafa, Michael R. Zile, and Romain Gallet
- Subjects
medicine.diagnostic_test ,Chemistry ,Kinase ,Diastole ,Exercise intolerance ,Pharmacology ,medicine.disease ,Preload ,Western blot ,Heart failure ,medicine ,medicine.symptom ,Protein kinase C ,PI3K/AKT/mTOR pathway - Abstract
IntroductionHeart failure with preserved ejection fraction (HFpEF) is characterized by diastolic dysfunction, pulmonary congestion and exercise intolerance. Previous preclinical studies show that treatment with cardiosphere-derived cells (CDCs) improves diastolic function, attenuates arrhythmias and prolongs survival in a rat HFpEF model. Here we characterize the myocardial proteome and diastolic function in HFpEF, with and without CDC therapy. As an initial strategy for identifying pathways worthy of further mechanistic dissection, we correlated CDC-responsive proteomic changes with functional improvements.Methods and ResultsDahl salt-sensitive rats fed high-salt diet, with verified diastolic dysfunction, were randomly assigned to intracoronary CDCs or placebo. Dahl rats fed a low salt diet served as controls. Phenotyping was by echocardiography (E/A ratio) and invasive hemodynamic monitoring (time constant of relaxation Tau, and left ventricular end-diastolic pressure [LVEDP]). CDC treatment improved diastolic function as indicated by a normalized E/A ratio, a 33.3% reduction in Tau, and a 47% reduction of LVEDP. Mass spectrometry of left ventricular tissues (n=6/group) revealed changes in transcription and translation pathways in this rat HFpEF model and was also recapitulated in human HFpEF. These pathways were enhanced following CDC treatment in the animal model (205 proteins and 32 phosphorylated residues accounting for 37% and 19% of all changes, respectively). Among all CDC-sensitive pathways, 65% can be linked to at least 1 of 7 upstream regulators, among which several are of potential relevance for regulating protein expression. To probe newly-synthesized proteins AHA labeling was carried out in isolated rat cardiomyocytes obtained from HFpEF groups, with and without CDC therapy. Five of the initial upstream regulators (HNF4A, MTOR, MYC, TGFβ1, and TP53) were linked to proteins expressed exclusively (or increased) with CDC treatment. All 32 phosphorylated residues of proteins involved in transcription/translation altered specifically by CDC treatment had predicted kinases (Protein kinase C (PKC) being the most dominant) and known to be regulated by MYC, TGFβ1 and/or TP53. Western blot analysis of those 5 upstream regulators showed that TGFβ1, TP53, and Myc were significantly decreased in LV from CDC treated animals, whereas MTOR and HNF4A showed a significant increase compared to HFpEF alone. The cellular quantities of several upstream regulator correlated with indices of diastolic function (E/A ratio, Tau and/or LVEDP). Since CDCs act via the secretion of exosomes laden with signaling cargo, it is relevant that all 7 upstream regulators could, in principle, be regulated by proteins or miRNA that are present in CDC-derived exosomes.ConclusionWe identified key cellular regulators of transcription and translation that underlie the therapeutic effects of CDCs in HFpEF, whose levels correlate quantitatively with measures of diastolic function. Among the multifarious proteomic changes associated with rat model of HFpEF which were also observed in human HFpEF samples, we propose that these regulators, and downstream effector kinases, be prioritized for further dedicated mechanistic dissection.
- Published
- 2021
8. PKG1-modified TSC2 regulates mTORC1 activity to counter adverse cardiac stress
- Author
-
Masayuki Sasaki, Christian U. Oeing, Dong Ik Lee, Djahida Bedja, Mark J. Ranek, Taishi Nakamura, Jonathan D. Powell, Jennifer E. Van Eyk, Chirag H. Patel, Anna Chen, Miguel Pinilla Vera, Ronald J. Holewinski, Kristen M. Kokkonen-Simon, Guangshuo Zhu, David A. Kass, and Brittany Dunkerly-Eyring
- Subjects
Male ,0301 basic medicine ,Heart Diseases ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,030204 cardiovascular system & hematology ,Article ,Mice ,Phosphoserine ,03 medical and health sciences ,0302 clinical medicine ,Tuberous Sclerosis Complex 2 Protein ,Autophagy ,Cyclic GMP-Dependent Protein Kinases ,Pressure ,Serine ,Animals ,Humans ,Myocytes, Cardiac ,Everolimus ,Gene Knock-In Techniques ,Phosphorylation ,Rats, Wistar ,Protein kinase A ,Mechanistic target of rapamycin ,Protein kinase B ,Cells, Cultured ,Pressure overload ,Multidisciplinary ,biology ,Kinase ,Chemistry ,Hypertrophy ,Rats ,Cell biology ,Enzyme Activation ,HEK293 Cells ,030104 developmental biology ,Mutation ,Disease Progression ,biology.protein ,Female ,biological phenomena, cell phenomena, and immunity ,RHEB - Abstract
The mechanistic target of rapamycin complex-1 (mTORC1) coordinates regulation of growth, metabolism, protein synthesis and autophagy1. 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
9. CHIP phosphorylation by protein kinase G enhances protein quality control and attenuates cardiac ischemic injury
- Author
-
Brittany Dunkerly-Eyring, Danielle Dillard, Masayuki Sasaki, Peter P. Rainer, Kristen M. Kokkonen-Simon, David A. Kass, Sumita Mishra, Richard C. Page, Jennifer E. Van Eyk, Virginia S. Hahn, Matthew M. Mannion, Huaqun Zhang, Christian U. Oeing, M. Imran Aslam, Mark J. Ranek, Ronald J. Holewinski, Jonathan C. Schisler, Dong I. Lee, Cornelia Virus, Monte S. Willis, Rebekah Sanchez-Hodge, and Vineet Agrawal
- Subjects
Male ,0301 basic medicine ,Science ,Ubiquitin-Protein Ligases ,Amino Acid Motifs ,General Physics and Astronomy ,macromolecular substances ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Ischemia ,Cyclic GMP-Dependent Protein Kinases ,Animals ,Humans ,Phosphorylation ,lcsh:Science ,Multidisciplinary ,biology ,Chemistry ,Myocardium ,Protein turnover ,Heart ,General Chemistry ,Cell biology ,Myocardial infarction ,Mechanisms of disease ,030104 developmental biology ,Proteostasis ,Proteasome ,Proteotoxicity ,Chaperone (protein) ,cardiovascular system ,biology.protein ,lcsh:Q ,Female ,cGMP-dependent protein kinase ,030217 neurology & neurosurgery - 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., Carboxyl terminus of Hsc70-interacting protein (CHIP) is proteostasis regulator. Here the authors show that CHIP-mediated protein turnover is enhanced by PKG-mediated phosphorylation, which results in attenuated cardiac ischemic proteotoxicity.
- Published
- 2020
10. Identification of Putative Early Atherosclerosis Biomarkers by Unsupervised Deconvolution of Heterogeneous Vascular Proteomes
- Author
-
Mitra Mastali, Jennifer E. Van Eyk, David M. Herrington, Georgia Saylor, Lulu Chen, Guoqiang Yu, Dana Troxlair, Ronald J. Holewinski, Weston R. Spivia, Chunhong Mao, Rakhi Pandey, Vidya Venkatraman, Richard S. Vander Heide, Grace Athas, Yue Wang, Sarah J. Parker, and Qin Fu
- Subjects
0301 basic medicine ,Proteomics ,Pathology ,medicine.medical_specialty ,Proteome ,Autopsy ,Coronary Artery Disease ,Anterior Descending Coronary Artery ,Biochemistry ,Article ,Lesion ,Coronary artery disease ,03 medical and health sciences ,Young Adult ,Medicine ,Humans ,030102 biochemistry & molecular biology ,Receiver operating characteristic ,business.industry ,Fatty streak ,General Chemistry ,medicine.disease ,Atherosclerosis ,030104 developmental biology ,Targeted mass spectrometry ,Female ,medicine.symptom ,business ,Biomarkers - Abstract
Coronary artery disease remains a leading cause of death in industrialized nations, and early detection of disease is a critical intervention target to effectively treat patients and manage risk. Proteomic analysis of mixed tissue homogenates may obscure subtle protein changes that occur uniquely in underlying tissue subtypes. The unsupervised 'convex analysis of mixtures' (CAM) tool has previously been shown to effectively segregate cellular subtypes from mixed expression data. In this study, we hypothesized that CAM would identify proteomic information specifically informative to early atherosclerosis lesion involvement that could lead to potential markers of early disease detection. We quantified the proteome of 99 paired abdominal aorta (AA) and left anterior descending coronary artery (LAD) specimens (N = 198 specimens total) acquired during autopsy of young adults free of diagnosed cardiac disease. The CAM tool was then used to segregate protein subsets uniquely associated with different underlying tissue types, yielding markers of normal and fibrous plaque (FP) tissues in LAD and AA (N = 62 lesions markers). CAM-derived FP marker expression was validated against pathologist estimated luminal surface involvement of FP, as well as in an orthogonal cohort of "pure" fibrous plaque, fatty streak, and normal vascular specimens. A targeted mass spectrometry (MS) assay quantified 39 of 62 CAM-FP markers in plasma from women with angiographically verified coronary artery disease (CAD, N = 46) or free from apparent CAD (control, N = 40). Elastic net variable selection with logistic regression reduced this list to 10 proteins capable of classifying CAD status in this cohort with
- Published
- 2020
11. A Dual Workflow to Improve the Proteomic Coverage in Plasma Using Data-Independent Acquisition-MS
- Author
-
Jennifer E. Van Eyk, Lilith Huang, Koen Raedschelders, Vidya Venkatraman, Qin Fu, Shenyan Zhang, and Ronald J. Holewinski
- Subjects
0301 basic medicine ,Detection limit ,Proteomics ,education.field_of_study ,030102 biochemistry & molecular biology ,biology ,Proteome ,Chemistry ,Population ,General Chemistry ,Computational biology ,Blood Proteins ,Mass spectrometry ,Biochemistry ,Blood proteins ,Mass Spectrometry ,Workflow ,03 medical and health sciences ,030104 developmental biology ,biology.protein ,Data-independent acquisition ,Antibody ,education ,Quantitative analysis (chemistry) - Abstract
Plasma is one of the most important and common matrices for clinical chemistry and proteomic analyses. Data-independent acquisition (DIA) mass spectrometry has enabled the simultaneous quantitative analysis of hundreds of proteins in plasma samples in support population and disease studies. Depletion of the highest abundant proteins is a common tool to increase plasma proteome coverage, but this strategy can result in the nonspecific depletion of protein subsets with which proteins targeted for depletion interact, adversely affecting their analysis. Our work using an antibody-based depletion column revealed significant complementarity not only in the identification of the proteins derived from depleted and undepleted plasma, but importantly also in the extent to which different proteins can be reproducibly quantified in each fraction. We systematically defined four major quantitative parameters of increasing stringency in both the depleted plasma fraction and in undepleted plasma for 757 observed plasma proteins: Linearity cutoff r2 > 0.8; lower limit of quantification (LLOQ); measurement range; limit of detection (LOD). We applied the results of our study to build a web-based tool, PlasmaPilot, that can serve as a protocol decision tree to determine whether the analysis of a specific protein warrants IgY14 mediated depletion.
- Published
- 2020
12. Quantitative Method for Assessing the Role of Lysine & Arginine Post-Translational Modifications in Nonalcoholic Steatohepatitis
- Author
-
Vidya Venkatraman, Ronald J. Holewinski, Peder J. Lund, Nathan Basisty, Aleksandra Binek, Xueshu Xie, Brian C. Searle, Aaron E. Robinson, Jennifer E. Van Eyk, Gautam Saxena, Birgit Schilling, George Rosenberger, Shelly C. Lu, Sarah J. Parker, Benjamin A. Garcia, and Jos eacute M Mato
- Subjects
chemistry.chemical_classification ,Succinylation ,Biochemistry ,Acetylation ,Chemistry ,DNA methylation ,Protein methylation ,Data-independent acquisition ,Peptide ,Multiplex ,Methylation - Abstract
Arg and Lys undergo multiple enzymatically driven post-translational modifications (PTMs), including methylation, which may be linked to key cellular processes. Today there is not a method that simultaneously quantifies proteins and the methylome at the site level. We have developed a method that can differentiate an unmodified peptide from its mono-, di- or tri-methylated Arg or Lys counterpart through a data independent acquisition approach that is suitable for both triple TOF and Orbitrap mass spectrometers. This method was further expanded to include Lys acetylation and succinylation, which in conjunction with methylation allow for simultaneous quantification of Lys PTMs in a single measurement. Our approach leverages small precursor mass windows to physically separate the various methylated species from each other during MS2 acquisition and shows linearity in the quantitation of low abundant peptides. We did this by first creating a biologically hyper-methylated peptide assay library, for mono-, di- and tri-methylated Lys and mono- and di-methylated Arg to which each experimental sample is compared. We further expanded the peptide assay library to include Lys acetylation and succinylation, providing the opportunity to multiplex five Lys and two Arg modification without the need for sample enrichment. To assess the validity of PTMs quantified with this method, we added an algorithm to determine the false localization rate for each potential modified amino acid residue. To evaluate its biological relevance, we applied this method to complex liver lysate from differentially methylated in-vivo non-alcoholic steatohepatitis mouse models and show that differential methylation potential altered not only protein quantity but also acetylation and to some degree succinylation. Of interest, acetylation data together with total protein changes drive strong novel hypothesis of a regulatory function of post-translational modifications in protein synthesis and mRNA stability. In conclusion: our method, which is suitable for different mass spectrometers along with the corresponding publicly available mouse liver PTM enriched peptide assay library, provides an easily adoptable framework needed for studying global protein methylation, acetylation, and succinylation in any proteomic experiment when total protein quantification is being carried out. Wide adoption of this easy approach will allow more rapid integration of PTM studies and knowledge.
- Published
- 2020
13. Protein S -Nitrosylation Controls Glycogen Synthase Kinase 3β Function Independent of Its Phosphorylation State
- Author
-
Zongming Fu, Vidya Venkatraman, Shengbing Wang, Mark J. Ranek, Ronald J. Holewinski, Jennifer E. Van Eyk, Ting Liu, David A. Kass, Erin L. Crowgey, and Brian O'Rourke
- Subjects
0301 basic medicine ,Physiology ,Chemistry ,Kinase ,HEK 293 cells ,S-Nitrosylation ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,GSK-3 ,Myocyte ,Phosphorylation ,Kinase activity ,Cardiology and Cardiovascular Medicine ,GSK3B ,030217 neurology & neurosurgery - Abstract
Rationale: GSK-3β (glycogen synthase kinase 3β) is a multifunctional and constitutively active kinase known to regulate a myriad of cellular processes. The primary mechanism to regulate its function is through phosphorylation-dependent inhibition at serine-9 residue. Emerging evidence indicates that there may be alternative mechanisms that control GSK-3β for certain functions. Objectives: Here, we sought to understand the role of protein S -nitrosylation (SNO) on the function of GSK-3β. SNO-dependent modulation of the localization of GSK-3β and its ability to phosphorylate downstream targets was investigated in vitro, and the network of proteins differentially impacted by phospho- or SNO-dependent GSK-3β regulation and in vivo SNO modification of key signaling kinases during the development of heart failure was also studied. Methods and Results: We found that GSK-3β undergoes site-specific SNO both in vitro, in HEK293 cells, H9C2 myoblasts, and primary neonatal rat ventricular myocytes, as well as in vivo, in hearts from an animal model of heart failure and sudden cardiac death. S -nitrosylation of GSK-3β significantly inhibits its kinase activity independent of the canonical phospho-inhibition pathway. S -nitrosylation of GSK-3β promotes its nuclear translocation and access to novel downstream phosphosubstrates which are enriched for a novel amino acid consensus sequence motif. Quantitative phosphoproteomics pathway analysis reveals that nuclear GSK-3β plays a central role in cell cycle control, RNA splicing, and DNA damage response. Conclusions: The results indicate that SNO has a differential effect on the location and activity of GSK-3β in the cytoplasm versus the nucleus. SNO modification of GSK-3β occurs in vivo and could contribute to the pathobiology of heart failure and sudden cardiac death.
- Published
- 2018
14. Application of volumetric absorptive microsampling for robust, high-throughput mass spectrometric quantification of circulating protein biomarkers
- Author
-
Stuart Kushon, Irene van den Broek, Michael P. Kowalski, Andrew Percy, Vidya Venkatraman, Ronald J. Holewinski, Qin Fu, Jennifer E. Van Eyk, and Kevin Millis
- Subjects
0301 basic medicine ,Reproducibility ,Chromatography ,medicine.diagnostic_test ,Protein biomarkers ,Chemistry ,010401 analytical chemistry ,Selected reaction monitoring ,Hematocrit ,Mass spectrometry ,Human serum albumin ,01 natural sciences ,0104 chemical sciences ,03 medical and health sciences ,030104 developmental biology ,medicine ,Sample preparation ,Data-independent acquisition ,Spectroscopy ,medicine.drug - Abstract
Volumetric absorptive micro sampling (VAMS™) allows accurate sampling of 10 µL of blood from a minimally invasive finger prick and could enable remote personalized health monitoring. Moreover, VAMS overcomes effects from hematocrit and sample heterogeneity associated with dried blood spots (DBS). We describe the first application of VAMS with the Mitra® microsampling device for the quantification of protein biomarkers using an automated, high-throughput sample preparation method coupled with mass spectrometric (MS) detection. The analytical performance of the developed workflow was evaluated for 10 peptides from six clinically relevant proteins: apolipoproteins A-I, B, C-I, C-III, E, and human serum albumin (HSA). Extraction recovery from blood with three different levels of hematocrit varied between 100% and 111% for all proteins. Within-day and total assay reproducibility (i.e., 5 replicates on 5 days) ranged between 3.2–10.4% and 3.4–12.6%, respectively. In addition, after 22 weeks of storage of the Mitra microsampling devices at −80 °C, all peptide responses were within ±15% deviation from the initial response. Application to data-independent acquisition (DIA) MS further demonstrated the potential for broad applicability and the general robustness of the automated workflow by reproducible detection of 1661 peptides from 423 proteins (average 15.7%CV (n = 3) in peptide abundance), correlating to peptide abundances in corresponding plasma (R = 0.8383). In conclusion, we have developed an automated workflow for efficient extraction, digestion, and MS analysis of a variety of proteins in a fixed small volume of dried blood (i.e., 10 µL). This robust and high-throughput workflow will create manifold opportunities for the application of remote, personalized disease biomarker monitoring.
- Published
- 2017
15. Mapping Citrullinated Sites in Multiple Organs of Mice Using Hypercitrullinated Library
- Author
-
Jennifer E. Van Eyk, Justyna Fert-Bober, Ronald J. Holewinski, Vidya Venkatraman, Aleksandr Stotland, Rakhi Pandey, Benjamin P. Berman, Ruining Liu, Erin L. Crowgey, and Christie L. Hunter
- Subjects
0301 basic medicine ,Male ,Computational biology ,Biology ,Kidney ,Biochemistry ,Protein citrullination ,Mass Spectrometry ,03 medical and health sciences ,Mice ,Peptide Library ,Animals ,Amino Acid Sequence ,Muscle, Skeletal ,Lung ,Regulation of gene expression ,030102 biochemistry & molecular biology ,Myocardium ,Citrullination ,Protein database ,Brain ,Computational Biology ,General Chemistry ,Mice, Inbred C57BL ,Subcellular distribution ,030104 developmental biology ,Liver ,Organ Specificity ,Proteome ,Posttranslational modification ,Research studies ,Protein-Arginine Deiminases ,Citrulline ,Muramidase ,Peptides ,Protein Processing, Post-Translational ,Metabolic Networks and Pathways ,Chromatography, Liquid - Abstract
Protein citrullination (or deimination), an irreversible post-translational modification, has been implicated in several physiological and pathological processes, including gene expression regulation, apoptosis, rheumatoid arthritis, and Alzheimer's disease. Several research studies have been carried out on citrullination under many conditions. However, until now, challenges in sample preparation and data analysis have made it difficult to confidently identify a citrullinated protein and assign the citrullinated site. To overcome these limitations, we generated a mouse hyper-citrullinated spectral library and set up coordinates to confidently identify and validate citrullinated sites. Using this workflow, we detect a four-fold increase in citrullinated proteome coverage across six mouse organs compared with the current state-of-the art techniques. Our data reveal that the subcellular distribution of citrullinated proteins is tissue-type-dependent and that citrullinated targets are involved in fundamental physiological processes, including the metabolic process. These data represent the first report of a hyper-citrullinated library for the mouse and serve as a central resource for exploring the role of citrullination in this organism.
- Published
- 2019
16. Protein kinase G signaling in cardiac pathophysiology: Impact of proteomics on clinical trials
- Author
-
Erin L. Crowgey, Jonathan A. Kirk, Jennifer E. Van Eyk, and Ronald J. Holewinski
- Subjects
0301 basic medicine ,Heart Diseases ,030204 cardiovascular system & hematology ,Biology ,Proteomics ,Biochemistry ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Cyclic GMP-Dependent Protein Kinases ,Animals ,Humans ,Amino Acid Sequence ,Cyclic GMP ,Molecular Biology ,Cyclic guanosine monophosphate ,Drug discovery ,Phosphodiesterase ,Heart ,Rats ,Cell biology ,030104 developmental biology ,Drug development ,chemistry ,cardiovascular system ,Phosphorylation ,Cattle ,Signal transduction ,cGMP-dependent protein kinase ,Signal Transduction - Abstract
The protective role of cyclic guanosine monophosphate (cGMP)-stimulated protein kinase G (PKG) in the heart makes it an attractive target for therapeutic drug development to treat a variety of cardiac diseases. Phosphodiesterases degrade cGMP, thus phosphodiesterase inhibitors that can increase PKG are of translational interest and the subject of ongoing human trials. PKG signaling is complex, however, and understanding its downstream phosphorylation targets and upstream regulation are necessary steps toward safe and efficacious drug development. Proteomic technologies have paved the way for assays that allow us to peer broadly into signaling minutia, including protein quantity changes and phosphorylation events. However, there are persistent challenges to the proteomic study of PKG, such as the impact of the expression of different PKG isoforms, changes in its localization within the cell, and alterations caused by oxidative stress. PKG signaling is also dependent upon sex and potentially the genetic and epigenetic background of the individual. Thus, the rigorous application of proteomics to the field will be necessary to address how these effectors can alter PKG signaling and interfere with pharmacological interventions. This review will summarize PKG signaling, how it is being targeted clinically, and the proteomic challenges and techniques that are being used to study it.
- Published
- 2016
17. Diabetes with heart failure increases methylglyoxal modifications in the sarcomere, which inhibit function
- Author
-
Samantha Beck Previs, Jennifer E. Van Eyk, Ronald J. Holewinski, Cheavar A. Blair, Thomas G Martin, Christine S. Moravec, Maria Papadaki, Jonathan A. Kirk, Marisa J. Stachowski, Kenneth S. Campbell, Amy Li, and David M. Warshaw
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Sarcomeres ,0301 basic medicine ,Myofilament ,medicine.medical_specialty ,Arginine ,Heart Ventricles ,Myosins ,030204 cardiovascular system & hematology ,Sarcomere ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Glycation ,Internal medicine ,Myosin ,medicine ,Animals ,Humans ,Myocyte ,Heart Failure ,Lysine ,Methylglyoxal ,General Medicine ,Middle Aged ,Pyruvaldehyde ,medicine.disease ,Actins ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,chemistry ,Heart failure ,Female ,Single-Cell Analysis ,Glycolysis - Abstract
Patients with diabetes are at significantly higher risk of developing heart failure. Increases in advanced glycation end products are a proposed pathophysiological link, but their impact and mechanism remain incompletely understood. Methylglyoxal (MG) is a glycolysis byproduct, elevated in diabetes, and modifies arginine and lysine residues. We show that left ventricular myofilament from patients with diabetes and heart failure (dbHF) exhibited increased MG modifications compared with nonfailing controls (NF) or heart failure patients without diabetes. In skinned NF human and mouse cardiomyocytes, acute MG treatment depressed both calcium sensitivity and maximal calcium-activated force in a dose-dependent manner. Importantly, dbHF myocytes were resistant to myofilament functional changes from MG treatment, indicating that myofilaments from dbHF patients already had depressed function arising from MG modifications. In human dbHF and MG-treated mice, mass spectrometry identified increased MG modifications on actin and myosin. Cosedimentation and in vitro motility assays indicate that MG modifications on actin and myosin independently depress calcium sensitivity, and mechanistically, the functional consequence requires actin/myosin interaction with thin-filament regulatory proteins. MG modification of the myofilament may represent a critical mechanism by which diabetes induces heart failure, as well as a therapeutic target to avoid the development of or ameliorate heart failure in these patients.
- Published
- 2018
18. Desmin Phosphorylation Triggers Preamyloid Oligomers Formation and Myocyte Dysfunction in Acquired Heart Failure
- Author
-
Yuchuan Wang, Martin G. Pomper, Peter P. Rainer, Nazareno Paolocci, Alessandra Baracca, Matteo Sorge, Gordon F. Tomaselli, Giulio Agnetti, Catherine A. Foss, Peihong Dong, Jennifer E. Van Eyk, Justyna Fert-Bober, Giancarlo Solaini, Ronald J. Holewinski, Steven S. An, Charles G. Glabe, Rainer, Peter P, Dong, Peihong, Sorge, Matteo, Fert-Bober, Justyna, Holewinski, Ronald J, Wang, Yuchuan, Foss, Catherine A, An, Steven S, Baracca, Alessandra, Solaini, Giancarlo, Glabe, Charles G, Pomper, Martin G, Van Eyk, Jennifer E, Tomaselli, Gordon F, Paolocci, Nazareno, and Agnetti, Giulio
- Subjects
Male ,0301 basic medicine ,Protein Folding ,Physiology ,Myocardial Ischemia ,heart failure ,030204 cardiovascular system & hematology ,Catechin ,Mass Spectrometry ,Mice ,0302 clinical medicine ,Myocyte ,Myocytes, Cardiac ,alpha-Crystallins ,Cytoskeleton ,Cells, Cultured ,Mice, Knockout ,Chemistry ,phosphorylation ,amyloid ,beta-Crystallins ,Recombinant Proteins ,Cell biology ,Phosphorylation ,Female ,Cardiology and Cardiovascular Medicine ,Amyloid ,Genetic Vectors ,desmin ,Polymorphism, Single Nucleotide ,Article ,Protein Aggregates ,03 medical and health sciences ,In vivo ,Pressure ,medicine ,Animals ,Humans ,proteostasis ,medicine.disease ,Rats ,030104 developmental biology ,Proteostasis ,Positron-Emission Tomography ,Heart failure ,Mutagenesis, Site-Directed ,bacteria ,Desmin ,Protein Processing, Post-Translational - Abstract
Rationale: Disrupted proteostasis is one major pathological trait that heart failure (HF) shares with other organ proteinopathies, such as Alzheimer and Parkinson diseases. Yet, differently from the latter, whether and how cardiac preamyloid oligomers (PAOs) develop in acquired forms of HF is unclear. Objective: We previously reported a rise in monophosphorylated, aggregate-prone desmin in canine and human HF. We now tested whether monophosphorylated desmin acts as the seed nucleating PAOs formation and determined whether positron emission tomography is able to detect myocardial PAOs in nongenetic HF. Methods and Results: Here, we first show that toxic cardiac PAOs accumulate in the myocardium of mice subjected to transverse aortic constriction and that PAOs comigrate with the cytoskeletal protein desmin in this well-established model of acquired HF. We confirm this evidence in cardiac extracts from human ischemic and nonischemic HF. We also demonstrate that Ser31 phosphorylated desmin aggregates extensively in cultured cardiomyocytes. Lastly, we were able to detect the in vivo accumulation of cardiac PAOs using positron emission tomography for the first time in acquired HF. Conclusions: Ser31 phosphorylated desmin is a likely candidate seed for the nucleation process leading to cardiac PAOs deposition. Desmin post-translational processing and misfolding constitute a new, attractive avenue for the diagnosis and treatment of the cardiac accumulation of toxic PAOs that can now be measured by positron emission tomography in acquired HF.
- Published
- 2018
19. Dephosphorylation of cardiac proteins in vitro - a matter of phosphatase specificity
- Author
-
Cathrine Husberg, Giulio Agnetti, Jennifer E. Van Eyk, Ronald J. Holewinski, Geir Christensen, Cathrine Husberg, Giulio Agnetti, Ronald J. Holewinski, Geir Christensen, and Jennifer E. Van Eyk
- Subjects
Proteomics ,biology ,Myocardium ,Phosphatase ,Muscle Proteins ,DUSP6 ,Protein phosphatase 1 ,heart ,Protein phosphatase 2 ,Biochemistry ,Article ,Cell biology ,Dephosphorylation ,Mice ,Protein Phosphatase 1 ,biology.protein ,Animals ,Alkaline phosphatase ,Phosphorylation ,Electrophoresis, Gel, Two-Dimensional ,Protein phosphorylation ,Protein Phosphatase 2 ,PHOSPHORYLATION ,Molecular Biology - Abstract
Protein phosphorylation is reversibly regulated by the interplay between kinases and phosphatases. Recent developments within the field of proteomics have revealed the extent of this modification in nature. To date there is still a lack of information about phosphatase specificity for different proteomes and their conditions to achieve maximum enzyme activity. This information is important per se, and in addition often requested in functional and biochemical in vitro studies, where a dephosphorylated sample is needed as a negative control to define baseline conditions. In this study, we have addressed the effectiveness of two phosphatases endogenously present in the heart (protein phosphatases 1 and 2A) and two generic phosphatases (alkaline phosphatase and lambda protein phosphatase) on three cardiac subproteomes known to be regulated by phosphorylation. We optimized the dephoshorylating conditions on a cardiac tissue fraction comprising cytosolic and myofilament proteins using 2DE and MS. The two most efficient conditions were further investigated on a mitochondrial-enriched fraction. Dephosphorylation of specific proteins depends on the phosphatase, its concentration, as well as sample preparation including buffer composition. Finally, we analyzed the efficiency of alkaline phosphatase, the phosphatase with the broadest substrate specificity, using TiO(2) peptide enrichment and 2DLC-MS/MS. Under these conditions, 95% of the detected cardiac cytoplasmic-enriched phospho-proteome was dephosphorylated. In summary, targeting dephosphorylation of the cardiac muscle subproteomes or a specific protein will drive the selection of the specific phosphatase, and each requires different conditions for optimal performance.
- Published
- 2012
20. Transient receptor potential channel 6 regulates abnormal cardiac S-nitrosylation in Duchenne muscular dystrophy
- Author
-
Ronald J. Holewinski, Jennifer E. Van Eyk, Djahida Bedja, David A. Kass, Guangshuo Zhu, Grace Kim, Heaseung Sophia Chung, and Vidya Venkatraman
- Subjects
0301 basic medicine ,Male ,Epinephrine ,Duchenne muscular dystrophy ,Nitrosation ,TRPC6 ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,medicine ,TRPC6 Cation Channel ,Animals ,Calcium Signaling ,Cysteine ,Sympathomimetics ,TRPC Cation Channels ,Multidisciplinary ,S-Nitrosothiols ,biology ,Ventricular Remodeling ,Chemistry ,Myocardium ,Cardiac muscle ,S-Nitrosylation ,medicine.disease ,Cell biology ,Nitric oxide synthase ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,biology.protein ,Mechanosensitive channels ,Dystrophin ,Gene Deletion - Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disorder with dystrophin loss that results in skeletal and cardiac muscle weakening and early death. Loss of the dystrophin-sarcoglycan complex delocalizes nitric oxide synthase (NOS) to alter its signaling, and augments mechanosensitive intracellular Ca2+ influx. The latter has been coupled to hyperactivation of the nonselective cation channel, transient receptor potential canonical channel 6 (Trpc6), in isolated myocytes. As Ca2+ also activates NOS, we hypothesized that Trpc6 would help to mediate nitric oxide (NO) dysregulation and that this would be manifest in increased myocardial S-nitrosylation, a posttranslational modification increasingly implicated in neurodegenerative, inflammatory, and muscle disease. Using a recently developed dual-labeling proteomic strategy, we identified 1,276 S-nitrosylated cysteine residues [S-nitrosothiol (SNO)] on 491 proteins in resting hearts from a mouse model of DMD (dmdmdx:utrn+/-). These largely consisted of mitochondrial proteins, metabolic regulators, and sarcomeric proteins, with 80% of them also modified in wild type (WT). S-nitrosylation levels, however, were increased in DMD. Genetic deletion of Trpc6 in this model (dmdmdx:utrn+/-:trpc6-/-) reversed ∼70% of these changes. Trpc6 deletion also ameliorated left ventricular dilation, improved cardiac function, and tended to reduce fibrosis. Furthermore, under catecholamine stimulation, which also increases NO synthesis and intracellular Ca2+ along with cardiac workload, the hypernitrosylated state remained as it did at baseline. However, the impact of Trpc6 deletion on the SNO proteome became less marked. These findings reveal a role for Trpc6-mediated hypernitrosylation in dmdmdx:utrn+/- mice and support accumulating evidence that implicates nitrosative stress in cardiac and muscle disease.
- Published
- 2017
21. Abstracts from the 8th International Conference on cGMP Generators, Effectors and Therapeutic Implications
- Author
-
Fritz G. Rathjen, Timothy D. Calamaras, Hong Wang, Motofumi Kumazoe, Sarah Helena Nies, Jerid W. Robinson, Andreas Friebe, Balázs Tamás Németh, Michel Negrerie, Kjetil W. Andressen, Clara D. Ledsky, Sivakanan Loganathan, Kerstin Jurk, Barbara Voussen, Julia Gorelik, Mi Kyung Chang, Johannes-Peter Stasch, Friederike Cuello, Nicholas C. Blixt, S. A. Dames, Nazareno Paolocci, Gzona Bajraktari, Katherine C. Hall, Markus Waldeck-Weiermair, Marcel A. Krüger, Antonius C.F. Gorren, Donté A. Stevens, Daniel Bloch, Deborah M. Dickey, Maria Hernandez-Valladares, Lan Zhao, Alex Vincent, Jenna Scotcher, Andrzej Weichsel, Tamás Radovits, Jeremy Richman, Sophie Schobesberger, Stavros Topouzis, Regine Mühlfriedel, Dennis Busse, Reiner Frey, Sean P. Parsons, René P. Zahedi, Evanthia Mergia, Mathias W. Seeliger, Angela McLaughlin, Sylvia Nikkho, Fernando Ferreira Costa, Pavel Jansa, Ornella Manfra, Wilson A. Ferreira, Inmaculada Silos-Santiago, B. Selin Kenet, Marc Humbert, Garyfalia Makrynitsa, Katja Stehfest, Lise Román Moltzau, Shiliang Li, Chris Sarko, Lincoln R. Potter, Yusu Gu, Scott A. Waldman, Nicole Eichert, David Kilpatrick, Roland Malli, Jun Hino, Robert Solinga, Banumathi Sankaran, William R. Montfort, Alexander Kollau, Julia Davydova, Andriana Stamopoulou, Olena Rudyk, Johanna Weiss, Kenji Kangawa, Arno Fritsch, Meinrad Gawaz, Peter Sandner, Robert M. Blanton, Friedrich Grimminger, Laurinda Jaffe, Atsuko Okamoto, Elke Butt, Stefanie Peters, Christine E. Gee, Ronald J. Holewinski, Zhi Cheng Jing, Andrea Gerling, Mika Takai, Alexander Froese, Hiroshi Hosoda, Mara Goetz, Yasutake Tanaka, Ioannis I. Alexandropoulos, Amie J Moyes, Hyazinth Dobrowinski, Michael J. Shipston, Ulrike Scheib, Stefan W. Hell, Oleksandra Prysyazhna, Lukas Rüttiger, Dieter Groneberg, Vu Thao-Vi Dao, Ulrich Walter, Hariharan Subramanian, Roger Flores-Costa, Richard C. Page, Joerg Reinders, Huaqun Zhang, Dianna M. Milewicz, Martin Thunemann, Albert T. Profy, Chieri Takeuchi, Andrew B. Tobin, Darren E. Casteel, Sarah Jacobson, Philip Eaton, Cor de Witt, Nadine Mauro, Mark Aronovitz, Sevil Korkmaz-Icöz, Barbara Wilhelm, Angelos Vachaviolos, Kim Tang, Axel Gödecke, Christian Meier, Carine M. Boustany, Daniel P. Zimmer, Bernd J. Pichler, Adrian J. Hobbs, Marianne Bjørnerem, Wolfgang F. Graier, Selene J. Sollie, Jenny Tobin, Jürgen Burhenne, Meinoshin Okumura, Nancy D. Dalton, Matthias Karck, Ernst-Martin Füchtbauer, Karoline Dröbner, Kristen M. Kokkonen, Flavia Menezes, Heribert Schunkert, Kirk L. Peterson, Ying-Ju Sung, Georgios A. Spyroulias, Olga N. Petrova, Frank Eitner, Mark G. Currie, Choel Kim, Ulrike Zimmermann, Steven S. Pullen, Shunhui Zhuang, Camila B. Almeida, Sylvie Bernier, M. Wittwer, Steffen Wolter, Stylianos Michalakis, Ralf Kühn, Sara S. O. Saad, Catherine Mansfield, Joon Jung, Jennifer E. Van Eyk, Navin K. Kapur, Renate B. Pilz, David Langleben, Diane Wong, Sylvie G. Bernier, Leia C. Shuhaibar, Heinz G. Körschen, César Ibarra-Alvarado, Courtney Shea, Ryan M. Fryer, Corina Russwurm, Gilbert W. Kim, Gerald Wölkart, Marius M. Hoeper, Astrid Weiss, Harald F. Langer, Ibrahim J. Domian, Lars I. Leichert, Bernd Wissinger, Paul Allan Renhowe, Richard H. Karas, Masayuki Sasaki, Finn Olav Levy, Cor de Wit, Sonia Donzelli, Michael Russwurm, Eberhart Zrenner, Reshma S. Baliga, Holly Clifford, Jonathan C. Schisler, Cheng-Yu Chen, Nicola Conran, Jens Schlossmann, Vikas Kapil, Pablo Colorado, Boris Tchernychev, Isabelle Lamarre, Katrin Reimann, Francheska Colon-Gonzalez, Ralph T. Schermuly, Hans-Ulrich Häring, Vineet Agrawal, Peter Germano, Bernd Mayer, Katharina Beck, Moritz Lehners, Sara Vandenwijngaert, Fabíola Z. Mónica, Pavel I. Nedvetsky, Kimberly Kafadar Long, Silja Meier, Paul A. Corris, Takeshi Tokudome, Shatanik Mukherjee, Joseph R. Burgoyne, Martin Deile, Jan Giesen, Ferid Murad, G. Todd Milne, Shinpei Kawaoka, Koichi Miura, Jessica A. Wales, David A. Kass, Doris Koesling, Hongxing Chen, Frode S. Berven, Jeong Joo Kim, Friedrich W. Herberg, Kristin Hartmann, Martin Biel, Raymond L. Benza, Monte S. Willis, Daniela Bertinetti, Jaime L. Masferrer, Stefan Z. Lutz, Guang-Rong Wang, Peter Ruth, Viacheslav O. Nikolaev, Tobias Peters, Karl Ulrich Bartz-Schmidt, Arnab Ghosh, Lai Wen, Mai Kadomatsu, Sandy M. Chu, Yutaka Suzuki, Marcus Olbrich, Yigao Huang, Tobias Bauch, Thorsten Kessler, Amrita Ahluwalia, Robert Feil, Daniel Richards, Hu Sheng Qian, M Currie, Christopher Newton-Cheh, V. Kaila, Liying Qin, Peter Hegemann, Peter Wright, G-Yoon Jamie Im, Hirofumi Tachibana, Kjestine Schmidt, Pamela L. Brito, Alisa Kamynina, James C. Campbell, Adam E. Snook, Fabian Schwiering, Martin Floor, Jordi Villà-Freixa, Jana Wobst, Alexandre Dumoulin, Ali R. Banijamali, Csaba Mátyás, Harald H.H.W. Schmidt, Susanne Kohl, Mark J. Ranek, Manuela Zaccolo, Robert Lukowski, Robert Naeije, Peter P. Rainer, Stephan Rosenkranz, Emmanuel S. Buys, Simone Romoli, Andreas Papapetropoulos, Kim C. Mansky, James Wakefield, Kálmán Benke, Takashi Nojiri, Aikaterini I. Argyriou, Dongjian Hu, Glenn A. Reinhart, Cornelia Virus, Marius Ueffing, Marlies Knipper, Gerhard Hannig, Kanako Takamatsu, Athanassios Giannis, Miki Arai, Stepan Gambaryan, James E. Sheppeck, Mathew M. Mannion, S. Bruce King, Henriette Andresen, Min Zhang, Jan D. Huizinga, Jeanette Erdmann, Matthias Broser, Astrid Schrammel, Dante J. Merlino, Hossein Ardeschir Ghofrani, Gaia Calamera, Obiajulu Agha, Kathleen A Lincoln, Gerburg K. Schwaerzer, Gordon L. Warren, Andrea Neubauer, Claire Poulet, Nicoletta C. Surdo, Attila Oláh, Damian Brockschnieder, Emrah Eroglu, Carla Fernanda Franco Penteado, Walter E. Haefeli, Mihály Ruppert, Ekkehard Grünig, Konstantina Stathopoulou, Elena Walter, Gérald Simonneau, Béla Merkely, Andreas Güldner, J Keppler, Katharina Frank, Renee Sarno, Sandra Frankenreiter, Sian E. Harding, Dorit Möhrle, Paul C. Harrison, Péter Hegedűs, Hyun-Soon Geisler, James R. Klinger, Markus Wolters, Philipp Henning, Jieru E. Lin, Erik S. Blomain, Alessandro Cataliotti, Ditta Zobor, Mikiya Miyazato, Marissa Opelt, Christian Schön, John Fassett, Yue Dai, Q. Luo, Thomas G. Oertner, Linda Breci, Robert A Baumgartner, Shun Hiroi, Ping Zhang, Albert Smolenski, Paul S. Frenette, Mahmood M. Alam, Jeremy R. Egbert, Dennis J. Stuehr, Eugen Franz, Hanan Chweih, Dong I. Lee, Nadja I. Bork, Guang Liu, Hannes Schmidt, Ka Bian, Annemarie Aue, Christian Dees, Edward Seto, Manuela Harloff, Michael Paolillo Hannes Schmidt, Susanne Feil, Susanne Krasemann, Andrea Hembre Ulsund, Marina Bantzi, Laurinda A. Jaffe, Luis Agulló, Dominik M. Fischer, Wendy Baur, Kjetil Wessel Andressen, Saurav Misra, Gábor Szabó, and Oliver Pagel
- Subjects
0301 basic medicine ,Pharmacology ,03 medical and health sciences ,030104 developmental biology ,business.industry ,Pharmacology toxicology ,MEDLINE ,Medicine ,Pharmacology (medical) ,business - Published
- 2017
22. Abstract 250: Canonical Transient Receptor Potential Channel 6 Ameliorates Increased Cardiac S-nitrosylation in Duchenne Muscular Dystrophy
- Author
-
Grace E Kim, Heaseung S Chung, Ronald J Holewinski, Guangshuo Zhu, Vidya Venkatraman, Djahida Bedja, Jennifer E Van Eyk, and David A Kass
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disorder that markedly weakens skeletal and cardiac muscle to cause early death. Its elimination of dystrophin disrupts nitric oxide (NO) signaling and amplifies intracellular Ca 2+ responses to mechanical load. We have shown the latter is linked to hyperstimulated transient receptor potential canonical 6 (TRPC6) cation channels. As Ca 2+ also activates NO synthase, we hypothesized TRPC6 couples to redox-dependent nitrosative stress to broadly impact protein S-nitrosylation (SNO). Using an unbiased, dual-labeling proteomic strategy we identified 1276 SNO sites on 491 proteins in DMD hearts (dystrophin/utrophin +/- ), of which 102 sites among 69 proteins were unique to DMD. Many of the targeted proteins were mitochondrial or metabolic regulators and sarcomere proteins - including titin, myosin binding protein-C, α-myosin heavy-chain, and tropomyosin α1 - that were hyper-nitrosylated. A key redox regulator peroxiredoxin1 was also hyper-nitrosylated at Cys173, a site previously shown to be a requisite regulator of its dimerization and enzymatic activity. DMD mice were then crossed into a Trpc6 -/- background, and proteomic analysis now found 70% of SNO targeted residues in DMD were reversed towards normal (p2 ). Trpc6 deletion improved left ventricular dilation (13.7±1.2mm, 22.4±3.9mm, 15.3±2.3mm; p-/- respectively (1-way ANOVA), and reversed pro-fibrotic gene activation (connective tissue growth factor, fibronectin1 and osteopontin). These results provide the first broad-based SNO analysis of the DMD heart, and support linkage between abnormal calcium via TRPC6, nitrosative stress and cardiac disease.
- Published
- 2017
23. Protein kinase A-dependent phosphorylation stimulates the transcriptional activity of hypoxia-inducible factor 1
- Author
-
Fan Wu, Irina Tchernyshyov, Jennifer E. Van Eyk, John W. Bullen, D. Kass, Robert N. Cole, Lauren R. DeVine, Ronald J. Holewinski, Gregg L. Semenza, and Vidya Venkatraman
- Subjects
0301 basic medicine ,Transcription, Genetic ,Allosteric regulation ,Biology ,GPI-Linked Proteins ,Biochemistry ,Hydroxylation ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Antigens, CD ,Gene expression ,Coactivator ,medicine ,Cyclic AMP ,Animals ,Humans ,Cyclic adenosine monophosphate ,Myocytes, Cardiac ,Phosphorylation ,Protein kinase A ,Molecular Biology ,5'-Nucleotidase ,Glutathione Transferase ,Immunosuppression Therapy ,Apyrase ,Cell Biology ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular biology ,Adenosine ,Cyclic AMP-Dependent Protein Kinases ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Disease Progression ,medicine.drug ,HeLa Cells ,Protein Binding - Abstract
Hypoxia-inducible factor 1 (HIF-1) activates the transcription of genes encoding proteins that enable cells to adapt to reduced O 2 availability. Proteins encoded by HIF-1 target genes play a central role in mediating physiological processes that are dysregulated in cancer and heart disease. These diseases are also characterized by increased production of cyclic adenosine monophosphate (cAMP), the allosteric activator of cAMP-dependent protein kinase A (PKA). Using glutathione S -transferase pull-down, coimmunoprecipitation, and mass spectrometry analyses, we demonstrated that PKA interacts with HIF-1α in HeLa cervical carcinoma cells and rat cardiomyocytes. PKA phosphorylated Thr 63 and Ser 692 on HIF-1α in vitro and enhanced HIF transcriptional activity and target gene expression in HeLa cells and rat cardiomyocytes. PKA inhibited the proteasomal degradation of HIF-1α in an O 2 -independent manner that required the phosphorylation of Thr 63 and Ser 692 and was not affected by prolyl hydroxylation. PKA also stimulated the binding of the coactivator p300 to HIF-1α to enhance its transcriptional activity and counteracted the inhibitory effect of asparaginyl hydroxylation on the association of p300 with HIF-1α. Furthermore, increased cAMP concentrations enhanced the expression of HIF target genes encoding CD39 and CD73, which are enzymes that convert extracellular adenosine 5′-triphosphate to adenosine, a molecule that enhances tumor immunosuppression and reduces heart rate and contractility. These data link stimuli that promote cAMP signaling, HIF-1α–dependent changes in gene expression, and increased adenosine, all of which contribute to the pathophysiology of cancer and heart disease.
- Published
- 2016
24. Methods for SWATH™: Data Independent Acquisition on TripleTOF Mass Spectrometers
- Author
-
Ronald J, Holewinski, Sarah J, Parker, Andrea D, Matlock, Vidya, Venkatraman, and Jennifer E, Van Eyk
- Subjects
Proteomics ,Humans ,Mass Spectrometry ,Software - Abstract
Data independent acquisition (DIA also termed SWATH) is an emerging technology in the field of mass spectrometry based proteomics. Although the concept of DIA has been around for over a decade, the recent advancements, in particular the speed of acquisition, of mass analyzers have pushed the technique into the spotlight and allowed for high-quality DIA data to be routinely acquired by proteomics labs. In this chapter we will discuss the protocols used for DIA acquisition using the Sciex TripleTOF mass spectrometers and data analysis using the Sciex processing software.
- Published
- 2016
25. Label-Free Quantification by Data Independent Acquisition Mass Spectrometry to Map Cardiovascular Proteomes
- Author
-
Vidya Venkatraman, Ronald J. Holewinski, Sarah J. Parker, Irina Tchernyshyov, Jennifer E. Van Eyk, and Laurie L. Parker
- Subjects
0301 basic medicine ,030102 biochemistry & molecular biology ,Computer science ,Proteomic Profiling ,Quantitative proteomics ,Computational biology ,Proteomics ,Mass spectrometry ,Bioinformatics ,03 medical and health sciences ,Label-free quantification ,030104 developmental biology ,Liquid chromatography–mass spectrometry ,Proteome ,Data-independent acquisition - Abstract
The large-scale identification and quantification of proteins by liquid chromatography mass spectrometry (LC MS) can be achieved by at least three general methods, categorized into targeted, data independent (DIA), and data dependent (DDA) acquisition modes. Each acquisition strategy has its own set of benefits and drawbacks, and the methods serve complementary purposes for the study of protein quantification in biological samples. While not specific to research in cardiovascular physiology, a long-standing but recently popularized proteomic approach, termed Data Independent Acquisition Mass Spectrometry (DIA-MS), promises unique strengths to complement and extend the existing capabilities of traditional “discovery” proteomic profiling by combining development of a peptide library and DIA-MS. In this chapter we will provide background on the DIA-MS technique, highlighting its fundamental differences relative to other mass spectrometry methods, and discuss important considerations for researchers interested in implementing this technique for their proteomic experiments.
- Published
- 2016
26. Methods for SWATH™: Data Independent Acquisition on TripleTOF Mass Spectrometers
- Author
-
Andrea Matlock, Sarah J. Parker, Vidya Venkatraman, Jennifer E. Van Eyk, and Ronald J. Holewinski
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,Mass spectrometry based proteomics ,Computer science ,Quantitative proteomics ,Data-independent acquisition ,Proteomics ,Bioinformatics ,Mass spectrometry ,Remote sensing - Abstract
Data independent acquisition (DIA also termed SWATH) is an emerging technology in the field of mass spectrometry based proteomics. Although the concept of DIA has been around for over a decade, the recent advancements, in particular the speed of acquisition, of mass analyzers have pushed the technique into the spotlight and allowed for high-quality DIA data to be routinely acquired by proteomics labs. In this chapter we will discuss the protocols used for DIA acquisition using the Sciex TripleTOF mass spectrometers and data analysis using the Sciex processing software.
- Published
- 2016
27. Pacemaker-induced transient asynchrony suppresses heart failure progression
- Author
-
Iraklis Pozios, Kyounghwan Lee, Gianluigi Pironti, Pieter P. de Tombe, Howard A. Rockman, Jonathan A. Kirk, Edward Karst, Taraneh Ghaffari Farazi, David A. Kass, Jennifer E. Van Eyk, Khalid Chakir, Roger Craig, Ronald J. Holewinski, Richard S. Tunin, and Theodore P. Abraham
- Subjects
Proteomics ,Sarcomeres ,Pacemaker, Artificial ,medicine.medical_specialty ,Myofilament ,Contraction (grammar) ,medicine.medical_treatment ,Cardiac resynchronization therapy ,Sarcomere ,Article ,Intracardiac injection ,Dogs ,Myofibrils ,Internal medicine ,Receptors, Adrenergic, beta ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,cardiovascular diseases ,Heart Failure ,Calcium metabolism ,business.industry ,General Medicine ,medicine.disease ,Heart failure ,Disease Progression ,cardiovascular system ,Cardiology ,Calcium ,business - Abstract
Uncoordinated contraction from electromechanical delay worsens heart failure pathophysiology and prognosis, but restoring coordination with biventricular pacing, known as cardiac resynchronization therapy (CRT), improves both. However, not every patient qualifies for CRT. We show that heart failure with synchronous contraction is improved by inducing dyssynchrony for 6 hours daily by right ventricular pacing using an intracardiac pacing device, in a process we call pacemaker-induced transient asynchrony (PITA). In dogs with heart failure induced by 6 weeks of atrial tachypacing, PITA (starting on week 3) suppressed progressive cardiac dilation as well as chamber and myocyte dysfunction. PITA enhanced β-adrenergic responsiveness in vivo and normalized it in myocytes. Myofilament calcium response declined in dogs with synchronous heart failure, which was accompanied by sarcomere disarray and generation of myofibers with severely reduced function, and these changes were absent in PITA-treated hearts. The benefits of PITA were not replicated when the same number of right ventricular paced beats was randomly distributed throughout the day, indicating that continuity of dyssynchrony exposure is necessary to trigger the beneficial biological response upon resynchronization. These results suggest that PITA could bring the benefits of CRT to the many heart failure patients with synchronous contraction who are not CRT candidates.
- Published
- 2015
28. Phospho-Proteomic Analysis of Cardiac Dyssynchrony and Resynchronization Therapy
- Author
-
Vidya Venkatraman, Eric Grote, Ronald J. Holewinski, Jonathan A. Kirk, Marisa J. Stachowski, and Jennifer E. Van Eyk
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Contraction (grammar) ,Proteome ,genetic structures ,medicine.medical_treatment ,Cardiac resynchronization therapy ,030204 cardiovascular system & hematology ,Biochemistry ,Article ,Cardiac Resynchronization Therapy ,03 medical and health sciences ,chemistry.chemical_compound ,Dogs ,0302 clinical medicine ,Western blot ,Tandem Mass Spectrometry ,Internal medicine ,Animals ,Medicine ,cardiovascular diseases ,Phosphorylation ,Molecular Biology ,Heart Failure ,Conduction abnormalities ,medicine.diagnostic_test ,business.industry ,Kinase ,Heart ,Tyrosine phosphorylation ,Phosphoproteins ,medicine.disease ,Treatment Outcome ,030104 developmental biology ,chemistry ,Heart failure ,cardiovascular system ,Cardiology ,business ,Biomarkers ,Signal Transduction - Abstract
Cardiac dyssynchrony arises from conduction abnormalities during heart failure and worsens morbidity and mortality. Cardiac Resynchronization Therapy (CRT) re-coordinates contraction using bi-ventricular pacing, but the cellular and molecular mechanisms involved remain largely unknown. We aimed to determine how dyssynchronous heart failure (HF(dys)) alters the phospho-proteome and how CRT interacts with this unique phospho-proteome by analyzing Ser/Thr and Tyr phosphorylation. Phospho-enriched myocardium from dog models of Control, HF(dys), and CRT was analyzed via mass spectrometry. There were 209 regulated phospho-sites among 1,761 identified sites. Compared to Con and CRT, HF(dys) was hyper-phosphorylated and tyrosine phosphorylation was more likely to be involved in signaling that increased with HF(dys) and was exacerbated by CRT. For each regulated site, the most-likely targeting-kinase was predicted, and CK2 was highly specific for sites that were “fixed” by CRT, suggesting activation of CK2 signaling occurs in HF(dys) that is reversed by CRT, which was supported by western blot analysis. These data elucidated signaling networks and kinases that may be involved and deserve further study. Importantly, we have identified a possible role for CK2 modulation in CRT. This may be harnessed in the future therapeutically to compliment CRT, improving its clinical effects.
- Published
- 2018
29. Newt cells secrete extracellular vesicles with therapeutic bioactivity in mammalian cardiomyocytes
- Author
-
Eleni Tseliou, Ryan Middleton, Kristin Luther, Eduardo Marbán, Geoffrey de Couto, Russell G. Rogers, Ronald J. Holewinski, Daniel Soetkamp, Travis J. Antes, and Jennifer E. Van Eyk
- Subjects
0301 basic medicine ,Histology ,Urodele amphibians ,cardiomyocytes ,tissue regeneration ,Biology ,Exosomes ,03 medical and health sciences ,0302 clinical medicine ,cytoprotection ,newt ,Secretion ,lcsh:QH573-671 ,Transcription factor ,PI3K/AKT/mTOR pathway ,lcsh:Cytology ,apoptosis ,RNA ,Cell Biology ,Microvesicles ,Cell biology ,PI3K/AKT pathway ,030104 developmental biology ,Nuclear receptor ,Apoptosis ,Signal transduction ,extracellular vesicles ,030217 neurology & neurosurgery ,Research Article - Abstract
Newts can regenerate amputated limbs and cardiac tissue, unlike mammals which lack broad regenerative capacity. Several signaling pathways involved in cell proliferation, differentiation and survival during newt tissue regeneration have been elucidated, however the factors that coordinate signaling between cells, as well as the conservation of these factors in other animals, are not well defined. Here we report that media conditioned by newt limb explant cells (A1 cells) protect mammalian cardiomyocytes from oxidative stress-induced apoptosis. The cytoprotective effect of A1-conditioned media was negated by exposing A1 cells to GW4869, which suppresses the generation of extracellular vesicles (EVs). A1-EVs are similar in diameter (~100–150 nm), structure, and share several membrane surface and cargo proteins with mammalian exosomes. However, isolated A1-EVs contain significantly higher levels of both RNA and protein per particle than mammalian EVs. Additionally, numerous cargo RNAs and proteins are unique to A1-EVs. Of particular note, A1-EVs contain numerous mRNAs encoding nuclear receptors, membrane ligands, as well as transcription factors. Mammalian cardiomyocytes treated with A1-EVs showed increased expression of genes in the PI3K/AKT pathway, a pivotal player in survival signaling. We conclude that newt cells secrete EVs with diverse, distinctive RNA and protein contents. Despite ~300 million years of evolutionary divergence between newts and mammals, newt EVs confer cytoprotective effects on mammalian cardiomyocytes.
- Published
- 2018
30. Methylglyoxal Modifications are Elevated in the Myofilament of Diabetic Cardiomyopathy Patients and Reduce Myofilament Function
- Author
-
Jonathan A. Kirk, Nikolai Smolin, Kenneth S. Campbell, Seth L. Robia, Ronald J. Holewinski, Maria Papadaki, Marisa J. Stachowski, and Cheavar A. Blair
- Subjects
medicine.medical_specialty ,Myofilament ,business.industry ,Methylglyoxal ,Biophysics ,medicine.disease ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Diabetic cardiomyopathy ,medicine ,Cardiology ,business ,Function (biology) - Published
- 2018
31. A novel phosphorylation site at Ser130 adjacent to the pseudosubstrate domain contributes to the activation of protein kinase C-δ
- Author
-
Jennifer E. Van Eyk, Susan F. Steinberg, Jianli Gong, and Ronald J. Holewinski
- Subjects
0301 basic medicine ,Allosteric regulation ,Molecular Sequence Data ,Biology ,Biochemistry ,Article ,MAP2K7 ,Substrate Specificity ,03 medical and health sciences ,Enzyme activator ,Serine ,Animals ,Humans ,Myocytes, Cardiac ,Amino Acid Sequence ,Kinase activity ,Phosphorylation ,Rats, Wistar ,Protein kinase A ,Molecular Biology ,Protein kinase C ,Kinase ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,Rats ,Enzyme Activation ,Protein Kinase C-delta ,030104 developmental biology ,Sequence Alignment - Abstract
Protein kinase C-δ (PKCδ) is a signalling kinase that regulates many cellular responses. Although most studies focus on allosteric mechanisms that activate PKCδ at membranes, PKCδ also is controlled via multi-site phosphorylation [Gong et al. (2015) Mol. Cell. Biol. 35: , 1727-1740]. The present study uses MS-based methods to identify PKCδ phosphorylation at Thr(50) and Ser(645) (in resting and PMA-treated cardiomyocytes) as well as Thr(37), Thr(38), Ser(130), Thr(164), Thr(211), Thr(215), Ser(218), Thr(295), Ser(299) and Thr(656) (as sites that increase with PMA). We focused on the consequences of phosphorylation at Ser(130) and Thr(141) (sites just N-terminal to the pseudosubstrate domain). We show that S130D and T141E substitutions co-operate to increase PKCδ's basal lipid-independent activity and that Ser(130)/Thr(141) di-phosphorylation influences PKCδ's substrate specificity. We recently reported that PKCδ preferentially phosphorylates substrates with a phosphoacceptor serine residue and that this is due to constitutive phosphorylation at Ser(357), an ATP-positioning G-loop site that limits PKCδ's threonine kinase activity [Gong et al. (2015) Mol. Cell. Biol. 35: , 1727-1740]. The present study shows that S130D and T141E substitutions increase PKCδ's threonine kinase activity indirectly by decreasing G loop phosphorylation at Ser(357). A S130F substitution [that mimics a S130F single-nt polymorphism (SNP) identified in some human populations] also increases PKCδ's maximal lipid-dependent catalytic activity and confers threonine kinase activity. Finally, we show that Ser(130)/Thr(141) phosphorylations relieve auto-inhibitory constraints that limit PKCδ's activity and substrate specificity in a cell-based context. Since phosphorylation sites map to similar positions relative to the pseudosubstrate domains of other PKCs, our results suggest that phosphorylation in this region of the enzyme may constitute a general mechanism to control PKC isoform activity.
- Published
- 2015
32. Posttranslational Modifications
- Author
-
Joel A. Kooren, Jennifer E. Van Eyk, Ronald J. Holewinski, and Justyna Fert-Bober
- Subjects
Chemistry ,Acetylation ,Protein phosphorylation ,Methylation ,Cell biology - Published
- 2015
33. Mass Spectrometry-Based Analysis of the Phospho-Proteome for Cardiac Dyssynchrony and Resynchronization Therapy
- Author
-
Ronald J. Holewinski, Jennifer E. Van Eyk, Jonathan A. Kirk, Eric Grote, David A. Kass, and Vidya Venkatraman
- Subjects
Cardiac function curve ,medicine.medical_specialty ,Phosphorylation sites ,Kinase ,business.industry ,medicine.medical_treatment ,Cardiac resynchronization therapy ,Biophysics ,medicine.disease ,Sarcomere ,Heart failure ,Internal medicine ,Proteome ,cardiovascular system ,medicine ,Cardiology ,business ,Tyrosine kinase - Abstract
In patients suffering from heart failure, conduction abnormalities that create cardiac dyssynchrony worsen morbidity and mortality. However, a pacemaker-based treatment termed Cardiac Resynchronization Therapy (CRT) can significantly restore function and mortality. Our previous work implicated post-translational signaling cascades in modulating this recovery. To further understand the mechanism of recovery, we aimed to catalogue, as completely as possible, the phospho-proteome in canine models of normal cardiac function (Con), cardiac dyssynchrony (Dys), and CRT. The models were generated by implanting pacemakers and pacing 6-weeks dyssynchronously (Dys), 3-weeks dyssynchronously and 3-weeks CRT (CRT), or not at all (Con). Each cardiac tissue sample (n=4 per group) was split into three parts: myofilament-enriched, myofilament-depleted, and phospho-tyrosine enriched. Each part was phospho-enriched and run on an Orbitrap Elite. By one-way ANOVA and Holm-Sidak post-hoc test, there were 104 sites Ser/Thr sites and 85 Tyr sites that were changed in at least one group. These sites were assigned one of three categories based on the pattern of changes: “Dyssynchrony, Not Fixed by CRT”, “Fixed by CRT”, or “CRT Only/Exacerbated by CRT”. Next we predicted the kinase for each phosphorylation site (Group-based Predication System 2.1). Many tyrosine kinases were identified, although no kinase showed specificity to any category. However, the Ser/Thr kinase CK2 was predicted to target a majority of the “Fixed By CRT” category, and was highly specific for this group, as CK2 was not predicted to target sites in any of the other categories. Thus, CRT appears to act as a broad CK2 activator. In the future, these sites need to be validated as CK2 targets. Moreover, the effect of CK2 activation on cardiomyocyte function, sarcomere shortening and calcium transient, will be explored, considering these are already implicated in recovery with CRT.
- Published
- 2015
- Full Text
- View/download PDF
34. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease
- Author
-
Djahida Bedja, Peter P. Rainer, Ronald J. Holewinski, Jennifer E. Van Eyk, Walter Paulus, Dong I. Lee, Manling Zhang, Jonathan A. Kirk, Kenneth B. Margulies, Thomas Danner, Takashi Sasaki, Eiki Takimoto, Nazha Hamdani, Wolfgang R. Dostmann, Mark J. Ranek, Su Hyun Jo, Guangshuo Zhu, Chulan Kwon, David A. Kass, Gun Sik Cho, Physiology, and ICaR - Heartfailure and pulmonary arterial hypertension
- Subjects
Male ,medicine.medical_specialty ,Phosphodiesterase Inhibitors ,Phosphodiesterase 3 ,Cardiomegaly ,030204 cardiovascular system & hematology ,Pharmacology ,Biology ,Nitric Oxide ,Nitric oxide ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Stress, Physiological ,Internal medicine ,Pressure ,medicine ,Animals ,Humans ,Natriuretic Peptides ,Cyclic GMP ,Cyclic guanosine monophosphate ,030304 developmental biology ,Muscle Cells ,0303 health sciences ,Multidisciplinary ,Myocardium ,Phosphodiesterase ,Aortic Valve Stenosis ,PDE5 drug design ,Up-Regulation ,3. Good health ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,3',5'-Cyclic-AMP Phosphodiesterases ,Nitric Oxide Pathway ,PDE10A ,Nitric Oxide Synthase ,cGMP-dependent protein kinase ,Signal Transduction - 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
35. The Role of Methylglyoxal on the Cardiac Myofilament
- Author
-
Jonathan A. Kirk, Maria Papadaki, Ronald J. Holewinski, and Marisa J. Stachowski
- Subjects
chemistry.chemical_compound ,Myofilament ,chemistry ,Methylglyoxal ,Biophysics ,Cell biology - Published
- 2017
36. Citrullination of myofilament proteins in heart failure
- Author
-
Erin L. Crowgey, Joan M. Bathon, Jonathan A. Kirk, John T. Giles, David A. Kass, Felipe Andrade, Jin Kyun Park, Justyna Fert-Bober, Helge Uhrigshardt, Marc K. Halushka, Jennifer E. Van Eyk, Clifton O. Bingham, and Ronald J. Holewinski
- Subjects
Gene isoform ,Male ,Myofilament ,Proteome ,Physiology ,Hydrolases ,Protein citrullination ,Mass Spectrometry ,chemistry.chemical_compound ,Myofibrils ,Physiology (medical) ,Protein-Arginine Deiminase Type 2 ,Myosin ,Citrulline ,Myocyte ,Animals ,Humans ,Adenosine Triphosphatases ,Heart Failure ,Myocardium ,Myosin Subfragments ,Citrullination ,Mice, Inbred C57BL ,Cytoskeletal Proteins ,chemistry ,Biochemistry ,Animals, Newborn ,Protein-Arginine Deiminases ,Cardiology and Cardiovascular Medicine - Abstract
Aims Citrullination, the post-translational conversion of arginine to citrulline by the enzyme family of peptidylarginine deiminases (PADs), is associated with several diseases, and specific citrullinated proteins have been shown to alter function while others act as auto-antigens. In this study, we identified citrullinated proteins in human myocardial samples, from healthy and heart failure patients, and determined several potential functional consequences. Further we investigated PAD isoform cell-specific expression in the heart. Methods and results A citrullination-targeted proteomic strategy using data-independent (SWATH) acquisition method was used to identify the modified cardiac proteins. Citrullinated-induced sarcomeric proteins were validated using two-dimensional gel electrophoresis and investigated using biochemical and functional assays. Myocardial PAD isoforms were confirmed by RT-PCR with PAD2 being the major isoform in myocytes. In total, 304 citrullinated sites were identified that map to 145 proteins among the three study groups: normal, ischaemia, and dilated cardiomyopathy. Citrullination of myosin (using HMM fragment) decreased its intrinsic ATPase activity and inhibited the acto-HMM-ATPase activity. Citrullinated TM resulted in stronger F-actin binding and inhibited the acto-HMM-ATPase activity. Citrullinated TnI did not alter the binding to F-actin or acto-HMM-ATPase activity. Overall, citrullination of sarcomeric proteins caused a decrease in Ca2+ sensitivity in skinned cardiomyocytes, with no change in maximal calcium-activated force or hill coefficient. Conclusion Citrullination unique to the cardiac proteome was identified. Our data indicate important structural and functional alterations to the cardiac sarcomere and the contribution of protein citrullination to this process.
- Published
- 2014
37. Cardiac resynchronization sensitizes the sarcomere to calcium by reactivating GSK-3?
- Author
-
Pieter P. de Tombe, David A. Kass, Giulio Agnetti, Jennifer E. Van Eyk, Namthip Witayavanitkul, Jonathan A. Kirk, Wei Dong Gao, Viola Kooij, Richard S. Tunin, Ronald J. Holewinski, Kirk JA, Holewinski RJ, Kooij V, Agnetti G, Tunin RS, Witayavanitkul N, de Tombe PP, Gao WD, Van Eyk J, and Kass DA
- Subjects
Sarcomeres ,medicine.medical_specialty ,Myofilament ,genetic structures ,medicine.medical_treatment ,Heart Ventricles ,Cardiac resynchronization therapy ,chemistry.chemical_element ,Biology ,Calcium ,Cell Enlargement ,In Vitro Techniques ,Sarcomere ,Glycogen Synthase Kinase 3 ,Dogs ,Myofibrils ,Troponin T ,Internal medicine ,Troponin I ,medicine ,Myocyte ,Animals ,cardiovascular diseases ,Phosphorylation ,Ventricular dyssynchrony ,Heart Failure ,General Medicine ,medicine.disease ,Myocardial Contraction ,Enzyme Activation ,Endocrinology ,chemistry ,Heart failure ,Cardiology ,cardiovascular system ,CARDIAC RESYNCHRONIZATION THERAPY ,Protein Processing, Post-Translational ,Research Article ,circulatory and respiratory physiology - Abstract
Cardiac resynchronization therapy (CRT), the application of biventricular stimulation to correct discoordinate contraction, is the only heart failure treatment that enhances acute and chronic systolic function, increases cardiac work, and reduces mortality. Resting myocyte function also increases after CRT despite only modest improvement in calcium transients, suggesting that CRT may enhance myofilament calcium responsiveness. To test this hypothesis, we examined adult dogs subjected to tachypacing-induced heart failure for 6 weeks, concurrent with ventricular dyssynchrony (HFdys) or CRT. Myofilament force-calcium relationships were measured in skinned trabeculae and/or myocytes. Compared with control, maximal calcium-activated force and calcium sensitivity declined globally in HFdys; however, CRT restored both. Phosphatase PP1 induced calcium desensitization in control and CRT-treated cells, while HFdys cells were unaffected, implying that CRT enhances myofilament phosphorylation. Proteomics revealed phosphorylation sites on Z-disk and M-band proteins, which were predicted to be targets of glycogen synthase kinase-3beta (GSK-3beta). We found that GSK-3beta was deactivated in HFdys and reactivated by CRT. Mass spectrometry of myofilament proteins from HFdys animals incubated with GSK-3beta confirmed GSK-3beta-dependent phosphorylation at many of the same sites observed with CRT. GSK-3beta restored calcium sensitivity in HFdys, but did not affect control or CRT cells. These data indicate that CRT improves calcium responsiveness of myofilaments following HFdys through GSK-3beta reactivation, identifying a therapeutic approach to enhancing contractile function.
- Published
- 2014
38. Characterization of the cardiac myosin binding protein-C phosphoproteome in healthy and failing human hearts
- Author
-
Viola Kooij, Ronald J. Holewinski, Anne M. Murphy, and Jennifer E. Van Eyk
- Subjects
Male ,Phosphorylation sites ,Proteome ,Muscle Proteins ,macromolecular substances ,Failing heart ,Biology ,Mice ,medicine ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Heart Failure ,Binding protein ,Myocardium ,Cardiac myosin ,medicine.disease ,Phosphoproteins ,Biochemistry ,Carrier protein ,Heart failure ,Female ,Cardiology and Cardiovascular Medicine ,Carrier Proteins ,Protein Kinases - Abstract
Cardiac myosin binding protein-C (cMyBP-C) becomes dephosphorylated in the failing heart and reduced phosphorylation-dependent regulation of cMyBP-C has been implicated in contractile dysfunction. To date, several phosphorylation sites have been identified for human cMyBP-C; however, a comprehensive characterization of the cMyBP-C phosphoproteome is lacking. This study aimed to characterize the cMyBP-C phosphoproteome using two different proteomic-based methods in explanted donor and end-stage failing hearts.The first approach used to characterize the cMyBP-C phosphoproteome employed a strong-cation exchange chromatography (SCX) fractionation method (10 pooled samples, technical replicates=4) and the second employed a sodium dodecylsulfate polyacrylamide gel electrophoresis method (n=10; technical replicates=2). Each subsequently underwent titanium dioxide (TiO2) affinity chromatography to enrich for the tryptic phosphopeptides, which were analyzed using an LTQ-Orbitrap mass spectrometer. Moreover, recombinant C0-C2 fragment of mouse cMyBP-C incubated with PKA, PKC, CaMKII and CK2 was analyzed to identify the kinases involved with phosphorylation of cMyBP-C.Seventeen phosphorylation sites on cMyBP-C were identified in vivo, with the majority localized in the N-terminal domains C0-C2. The three most abundant phosphorylated sites, Ser284, Ser286 and Thr290, are located in the regulatory M-domain of cMyBP-C. Ser284 showed a significant reduction in phosphorylation in HF.This study demonstrates that cMyBP-C harbors more phosphorylation sites than previously known, with a total of 17 (9 novel) identified phosphorylation sites in vivo. Most sites were primarily located within the N-terminal side of the protein. The most highly phosphorylated site on cMyBP-C was Ser284 and this site showed decreased phosphorylation in the failing heart, which implicates importance for fine-tuning contractility. To date, the functional importance of Ser286 and Thr290 is unknown. In addition, 16 sites were identified after in vitro kinase incubation. The data have been deposited to the ProteomeXchange with identifier PXD000158.
- Published
- 2013
39. A fast and reproducible method for albumin isolation and depletion from serum and cerebrospinal fluid
- Author
-
Matthew J. Powell, Zhicheng Jin, Ronald J. Holewinski, Jennifer E. Van Eyk, and Matthew D. Maust
- Subjects
Proteomics ,Chromatography ,Proteome ,Blotting, Western ,Albumin ,Reproducibility of Results ,Biology ,Tandem mass spectrometry ,Biochemistry ,Article ,Cerebrospinal fluid ,Tandem Mass Spectrometry ,Albumins ,biology.protein ,Biomarker (medicine) ,Humans ,Electrophoresis, Polyacrylamide Gel ,Biomarker discovery ,Bovine serum albumin ,Molecular Biology ,Biomarkers ,Chromatography, Liquid - Abstract
Analysis of serum and plasma proteomes is a common approach for biomarker discovery, and the removal of high abundant proteins, such as albumin and immunoglobins, is usually the first step in the analysis of serum and plasma proteomes. However, albumin binds peptides and proteins, which raises concerns as to how the removal of albumin could impact the outcome of the biomarker study while ignoring the possibility that this could be a biomarker sub-proteome itself. The first goal of this study was to test a new commercially available affinity capture reagent from Protea Biosciences and to compare the efficiency and reproducibility to 4 other commercially available albumin depletion methods. The second goal of this study was to determine if there is a highly efficient albumin depletion/isolation system that minimizes sample handling and would be suitable for large numbers of samples. Two of the methods tested (Sigma and ProteaPrep) showed an albumin depletion efficiency of 97% or greater for both serum and cerebrospinal fluid (CSF). Isolated serum and CSF albuminomes from ProteaPrep spin columns were analyzed directly by LC-MS/MS, identifying 128 serum (45 not previously reported) and 94 CSF albuminome proteins (17 unique to the CSF albuminome). Serum albuminome was also isolated using Vivapure anti-HSA columns for comparison, identifying 105 proteins, 81 of which overlapped with the ProteaPrep method.
- Published
- 2012
40. Inhibition by active site directed covalent modification of human glyoxalase I
- Author
-
Ronald J. Holewinski and Donald J. Creighton
- Subjects
Inhibitor ,GLO1 ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Structure-Activity Relationship ,Lactoylglutathione lyase ,chemistry.chemical_compound ,Glycation ,Catalytic Domain ,Drug Discovery ,Humans ,Advanced glycation end-products (AGEs) ,Enzyme Inhibitors ,Molecular Biology ,Glyoxalase ,Medicine(all) ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Covalent modification ,Organic Chemistry ,Methylglyoxal ,Lactoylglutathione Lyase ,Active site ,Glutathione ,Inactivator ,In vitro ,Enzyme ,chemistry ,biology.protein ,Molecular Medicine ,Intracellular ,DNA - Abstract
The glyoxalase pathway is responsible for conversion of cytotoxic methylglyoxal (MG) to d-lactate. MG toxicity arises from its ability to form advanced glycation end products (AGEs) on proteins, lipids and DNA. Studies have shown that inhibitors of glyoxalase I (GLO1), the first enzyme of this pathway, have chemotherapeutic effects both in vitro and in vivo, presumably by increasing intracellular MG concentrations leading to apoptosis and cell death. Here, we present the first molecular inhibitor, 4-bromoacetoxy-1-(S-glutathionyl)-acetoxy butane (4BAB), able to covalently bind to the free sulfhydryl group of Cys60 in the hydrophobic binding pocket adjacent to the enzyme active site and partially inactivate the enzyme. Our data suggests that partial inactivation of homodimeric GLO1 is due to the modification at only one of the enzymatic active sites. Although this molecule may have limited use pharmacologically, it may serve as an important template for the development of new GLO1 inhibitors that may combine this strategy with ones already reported for high affinity GLO1 inhibitors, potentially improving potency and specificity.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.