369 results on '"Monte S. Willis"'
Search Results
2. CHIP phosphorylation by protein kinase G enhances protein quality control and attenuates cardiac ischemic injury
- Author
-
Mark J. Ranek, Christian Oeing, Rebekah Sanchez-Hodge, Kristen M. Kokkonen-Simon, Danielle Dillard, M. Imran Aslam, Peter P. Rainer, Sumita Mishra, Brittany Dunkerly-Eyring, Ronald J. Holewinski, Cornelia Virus, Huaqun Zhang, Matthew M. Mannion, Vineet Agrawal, Virginia Hahn, Dong I. Lee, Masayuki Sasaki, Jennifer E. Van Eyk, Monte S. Willis, Richard C. Page, Jonathan C. Schisler, and David A. Kass
- Subjects
Science - Abstract
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
- Full Text
- View/download PDF
3. Atrogin-1 inhibits Akt-dependent cardiac hypertrophy in mice via ubiquitin-dependent coactivation of Forkhead proteins
- Author
-
Hui-Hua Li, Monte S. Willis, Pamela Lockyer, Nathaniel Miller, Holly McDonough, David J. Glass, and Cam Patterson
- Subjects
Medicine - Published
- 2022
- Full Text
- View/download PDF
4. Cardiomyocyte microRNA-150 confers cardiac protection and directly represses proapoptotic small proline–rich protein 1A
- Author
-
Tatsuya Aonuma, Bruno Moukette, Satoshi Kawaguchi, Nipuni P. Barupala, Marisa N. Sepúlveda, Christopher Corr, Yaoliang Tang, Suthat Liangpunsakul, R. Mark Payne, Monte S. Willis, and Il-man Kim
- Subjects
Cardiology ,Cell biology ,Medicine - Abstract
MicroRNA-150 (miR-150) is downregulated in patients with multiple cardiovascular diseases and in diverse mouse models of heart failure (HF). miR-150 is significantly associated with HF severity and outcome in humans. We previously reported that miR-150 is activated by β-blocker carvedilol (Carv) and plays a protective role in the heart using a systemic miR-150 KO mouse model. However, mechanisms that regulate cell-specific miR-150 expression and function in HF are unknown. Here, we demonstrate that potentially novel conditional cardiomyocyte–specific (CM-specific) miR-150 KO (miR-150 cKO) in mice worsens maladaptive cardiac remodeling after myocardial infarction (MI). Genome-wide transcriptomic analysis in miR-150 cKO mouse hearts identifies small proline–rich protein 1a (Sprr1a) as a potentially novel target of miR-150. Our studies further reveal that Sprr1a expression is upregulated in CMs isolated from ischemic myocardium and subjected to simulated ischemia/reperfusion, while its expression is downregulated in hearts and CMs by Carv. We also show that left ventricular SPRR1A is upregulated in patients with HF and that Sprr1a knockdown in mice prevents maladaptive post-MI remodeling. Lastly, protective roles of CM miR-150 are, in part, attributed to the direct and functional repression of proapoptotic Sprr1a. Our findings suggest a crucial role for the miR-150/SPRR1A axis in regulating CM function post-MI.
- Published
- 2021
- Full Text
- View/download PDF
5. The Access Technology Program of the Indiana Clinical Translational Sciences Institute (CTSI): A model to facilitate access to cutting-edge technologies across a state
- Author
-
Christie M. Orschell, Todd C. Skaar, Melanie E. DeFord, Joel Ybe, Julie Driscol, Christine Drury, Lilith Reeves, Monte S. Willis, Jill L. Reiter, Jenna York, Rob Orr, Jeanette N. McClintick, Thomas G. Sors, Joe Hunt, Kenneth Cornetta, and Anantha Shekhar
- Subjects
Translational research ,technology ,CTSI ,core facilities ,pilot funding ,Medicine - Abstract
Abstract Introduction: Access to cutting-edge technologies is essential for investigators to advance translational research. The Indiana Clinical and Translational Sciences Institute (CTSI) spans three major and preeminent universities, four large academic campuses across the state of Indiana, and is mandate to provide best practices to a whole state. Methods: To address the need to facilitate the availability of innovative technologies to its investigators, the Indiana CTSI implemented the Access Technology Program (ATP). The activities of the ATP, or any program of the Indiana CTSI, are challenged to connect technologies and investigators on the multiple Indiana CTSI campuses by the geographical distances between campuses (1–4 hr driving time). Results: Herein, we describe the initiatives developed by the ATP to increase the availability of state-of-the-art technologies to its investigators on all Indiana CTSI campuses, and the methods developed by the ATP to bridge the distance between campuses, technologies, and investigators for the advancement of clinical translational research. Conclusions: The methods and practices described in this publication may inform other approaches to enhance translational research, dissemination, and usage of innovative technologies by translational investigators, especially when distance or multi-campus cultural differences are factors to efficient application.
- Published
- 2021
- Full Text
- View/download PDF
6. Muscle-specific regulation of right ventricular transcriptional responses to chronic hypoxia-induced hypertrophy by the muscle ring finger-1 (MuRF1) ubiquitin ligase in mice
- Author
-
Robert H. Oakley, Matthew J. Campen, Michael L. Paffett, Xin Chen, Zhongjing Wang, Traci L. Parry, Carolyn Hillhouse, John A. Cidlowski, and Monte S. Willis
- Subjects
MuRF1 ,Hypoxia ,Right heart failure ,Gene expression ,Microarray ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background We recently identified a role for the muscle-specific ubiquitin ligase MuRF1 in right-sided heart failure secondary to pulmonary hypertension induced by chronic hypoxia (CH). MuRF1−/− mice exposed to CH are resistant to right ventricular (RV) dysfunction whereas MuRF1 Tg + mice exhibit impaired function indicative of heart failure. The present study was undertaken to understand the underlying transcriptional alterations in the RV of MuRF1−/− and MuRF1 Tg + mice. Methods Microarray analysis was performed on RNA isolated from the RV of MuRF1−/−, MuRF1 Tg+, and wild-type control mice exposed to CH. Results MuRF1−/− RV differentially expressed 590 genes in response to CH. Analysis of the top 66 genes (> 2-fold or 3-fold or
- Published
- 2018
- Full Text
- View/download PDF
7. 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
8. Modeling the Transition From Decompensated to Pathological Hypertrophy
- Author
-
Florencia Pascual, Jonathan C. Schisler, Trisha J. Grevengoed, Monte S. Willis, and Rosalind A. Coleman
- Subjects
fatty acid ,fibrosis ,fuel switching ,glycolysis ,metabolomics ,mTOR ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
BackgroundLong‐chain acyl‐CoA synthetases (ACSL) catalyze the conversion of long‐chain fatty acids to fatty acyl‐CoAs. Cardiac‐specific ACSL1 temporal knockout at 2 months results in a shift from FA oxidation toward glycolysis that promotes mTORC1‐mediated ventricular hypertrophy. We used unbiased metabolomics and gene expression analyses to examine the early effects of genetic inactivation of fatty acid oxidation on cardiac metabolism, hypertrophy development, and function. Methods and ResultsGlobal cardiac transcriptional analysis revealed differential expression of genes involved in cardiac metabolism, fibrosis, and hypertrophy development in Acsl1H−/− hearts 2 weeks after Acsl1 ablation. Comparison of the 2‐ and 10‐week transcriptional responses uncovered 137 genes whose expression was uniquely changed upon knockdown of cardiac ACSL1, including the distinct upregulation of fibrosis genes, a phenomenon not observed after complete ACSL1 knockout. Metabolomic analysis identified metabolites altered in hearts displaying partially reduced ACSL activity, and rapamycin treatment normalized the cardiac metabolomic fingerprint. ConclusionsShort‐term cardiac‐specific ACSL1 inactivation resulted in metabolic and transcriptional derangements distinct from those observed upon complete ACSL1 knockout, suggesting heart‐specific mTOR (mechanistic target of rapamycin) signaling that occurs during the early stages of substrate switching. The hypertrophy observed with partial Acsl1 ablation occurs in the context of normal cardiac function and is reminiscent of a physiological process, making this a useful model to study the transition from physiological to pathological hypertrophy.
- Published
- 2018
- Full Text
- View/download PDF
9. Kinome and Transcriptome Profiling Reveal Broad and Distinct Activities of Erlotinib, Sunitinib, and Sorafenib in the Mouse Heart and Suggest Cardiotoxicity From Combined Signal Transducer and Activator of Transcription and Epidermal Growth Factor Receptor Inhibition
- Author
-
Timothy J. Stuhlmiller, Jon S. Zawistowski, Xin Chen, Noah Sciaky, Steven P. Angus, Sean T. Hicks, Traci L. Parry, Wei Huang, Ju Youn Beak, Monte S. Willis, Gary L. Johnson, and Brian C. Jensen
- Subjects
antineoplastic agents ,cardiomyopathy ,cardiotoxicity ,protein kinase inhibitors ,proteomics ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
BackgroundMost novel cancer therapeutics target kinases that are essential to tumor survival. Some of these kinase inhibitors are associated with cardiotoxicity, whereas others appear to be cardiosafe. The basis for this distinction is unclear, as are the molecular effects of kinase inhibitors in the heart. Methods and ResultsWe administered clinically relevant doses of sorafenib, sunitinib (cardiotoxic multitargeted kinase inhibitors), or erlotinib (a cardiosafe epidermal growth factor receptor inhibitor) to mice daily for 2 weeks. We then compared the effects of these 3 kinase inhibitors on the cardiac transcriptome using RNAseq and the cardiac kinome using multiplexed inhibitor beads coupled with mass spectrometry. We found unexpectedly broad molecular effects of all 3 kinase inhibitors, suggesting that target kinase selectivity does not define either the molecular response or the potential for cardiotoxicity. Using in vivo drug administration and primary cardiomyocyte culture, we also show that the cardiosafety of erlotinib treatment may result from upregulation of the cardioprotective signal transducer and activator of transcription 3 pathway, as co‐treatment with erlotinib and a signal transducer and activator of transcription inhibitor decreases cardiac contractile function and cardiomyocyte fatty acid oxidation. ConclusionsCollectively our findings indicate that preclinical kinome and transcriptome profiling may predict the cardiotoxicity of novel kinase inhibitors, and suggest caution for the proposed therapeutic strategy of combined signal transducer and activator of transcription/epidermal growth factor receptor inhibition for cancer treatment.
- Published
- 2017
- Full Text
- View/download PDF
10. MMI‐0100 Inhibits Cardiac Fibrosis in a Mouse Model Overexpressing Cardiac Myosin Binding Protein C
- Author
-
Qinghang Meng, Bidur Bhandary, Hanna Osinska, Jeanne James, Na Xu, Kritton Shay‐Winkler, James Gulick, Monte S. Willis, Cynthia Lander, and Jeffrey Robbins
- Subjects
fibrosis ,hypertrophy/remodeling ,transgenic mice ,transgenic model ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
BackgroundCardiac stress can trigger production of a 40‐kDa peptide fragment derived from the amino terminus of the cardiac myosin‐binding protein C. Cardiac stress, as well as cMyBP‐C mutations, can trigger production of 1 such truncated protein fragment, a 40‐kDa peptide fragment derived from the amino terminus of cMyBP‐C. Genetic expression of this 40‐kDa fragment in mouse cardiomyocytes (cMyBP‐C40k) leads to cardiac disease, fibrosis, and death within the first year. Fibrosis can occur in many cardiovascular diseases, and mitogen‐activated protein kinase––activated protein kinase‐2 signaling has been implicated in a variety of fibrotic processes. Recent studies demonstrated that mitogen‐activated protein kinase––activated protein kinase‐2 inhibition using the cell‐permeant peptide inhibitor MMI‐0100 is protective in the setting of acute myocardial infarction. We hypothesized that MMI‐0100 might also be protective in a chronic model of fibrosis, produced as a result of cMyBP‐C40k cardiomyocyte expression. Methods and ResultsNontransgenic and cMyBP‐C40k inducible transgenic mice were given MMI‐0100 or PBS daily for 30 weeks. In control groups, long‐term MMI‐0100 was benign, with no measurable effects on cardiac anatomy, function, cell viability, hypertrophy, or probability of survival. In the inducible transgenic group, MMI‐0100 treatment reduced cardiac fibrosis, decreased cardiac hypertrophy, and prolonged survival. ConclusionsPharmaceutical inhibition of mitogen‐activated protein kinase––activated protein kinase‐2 signaling via MMI‐0100 treatment is beneficial in the context of fibrotic cMyBPC40k disease.
- Published
- 2017
- Full Text
- View/download PDF
11. Adverse Effects of Fenofibrate in Mice Deficient in the Protein Quality Control Regulator, CHIP
- Author
-
Saranya Ravi, Traci L. Parry, Monte S. Willis, Pamela Lockyer, Cam Patterson, James R. Bain, Robert D. Stevens, Olga R. Ilkayeva, Christopher B. Newgard, and Jonathan C. Schisler
- Subjects
metabolism ,fibrates ,fibrosis ,metabolomics ,pressure overload ,autophagy ,mitophagy ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
We previously reported how the loss of CHIP expression (Carboxyl terminus of Hsc70-Interacting Protein) during pressure overload resulted in robust cardiac dysfunction, which was accompanied by a failure to maintain ATP levels in the face of increased energy demand. In this study, we analyzed the cardiac metabolome after seven days of pressure overload and found an increase in long-chain and medium-chain fatty acid metabolites in wild-type hearts. This response was attenuated in mice that lack expression of CHIP (CHIP−/−). These findings suggest that CHIP may play an essential role in regulating oxidative metabolism pathways that are regulated, in part, by the nuclear receptor PPARα (Peroxisome Proliferator-Activated Receptor alpha). Next, we challenged CHIP−/− mice with the PPARα agonist called fenofibrate. We found that treating CHIP−/− mice with fenofibrate for five weeks under non-pressure overload conditions resulted in decreased skeletal muscle mass, compared to wild-type mice, and a marked increase in cardiac fibrosis accompanied by a decrease in cardiac function. Fenofibrate resulted in decreased mitochondrial cristae density in CHIP−/− hearts as well as decreased expression of genes involved in the initiation of autophagy and mitophagy, which suggests that a metabolic challenge, in the absence of CHIP expression, impacts pathways that contribute to mitochondrial quality control. In conclusion, in the absence of functional CHIP expression, fenofibrate results in unexpected skeletal muscle and cardiac pathologies. These findings are particularly relevant to patients harboring loss-of-function mutations in CHIP and are consistent with a prominent role for CHIP in regulating cardiac metabolism.
- Published
- 2018
- Full Text
- View/download PDF
12. Exercise-Induced Alterations in Skeletal Muscle, Heart, Liver, and Serum Metabolome Identified by Non-Targeted Metabolomics Analysis
- Author
-
Joseph W. Starnes, Traci L. Parry, Sara K. O’Neal, James R. Bain, Michael J. Muehlbauer, Aubree Honcoop, Amro Ilaiwy, Peter M. Christopher, Cam Patterson, and Monte S. Willis
- Subjects
exercise ,metabolism ,skeletal muscle ,heart ,liver ,serum ,non-targeted metabolomics ,Microbiology ,QR1-502 - Abstract
Background: The metabolic and physiologic responses to exercise are increasingly interesting, given that regular physical activity enhances antioxidant capacity, improves cardiac function, and protects against type 2 diabetes. The metabolic interactions between tissues and the heart illustrate a critical cross-talk we know little about. Methods: To better understand the metabolic changes induced by exercise, we investigated skeletal muscle (plantaris, soleus), liver, serum, and heart from exercise trained (or sedentary control) animals in an established rat model of exercise-induced aerobic training via non-targeted GC-MS metabolomics. Results: Exercise-induced alterations in metabolites varied across tissues, with the soleus and serum affected the least. The alterations in the plantaris muscle and liver were most alike, with two metabolites increased in each (citric acid/isocitric acid and linoleic acid). Exercise training additionally altered nine other metabolites in the plantaris (C13 hydrocarbon, inosine/adenosine, fructose-6-phosphate, glucose-6-phosphate, 2-aminoadipic acid, heptadecanoic acid, stearic acid, alpha-tocopherol, and oleic acid). In the serum, we identified significantly decreased alpha-tocopherol levels, paralleling the increases identified in plantaris muscle. Eleven unique metabolites were increased in the heart, which were not affected in the other compartments (malic acid, serine, aspartic acid, myoinositol, glutamine, gluconic acid-6-phosphate, glutamic acid, pyrophosphate, campesterol, phosphoric acid, creatinine). These findings complement prior studies using targeted metabolomics approaches to determine the metabolic changes in exercise-trained human skeletal muscle. Specifically, exercise trained vastus lateralus biopsies had significantly increased linoleic acid, oleic acid, and stearic acid compared to the inactive groups, which were significantly increased in plantaris muscle in the present study. Conclusions: While increases in alpha-tocopherol have not been identified in muscle after exercise to our knowledge, the benefits of vitamin E (alpha-tocopherol) supplementation in attenuating exercise-induced muscle damage has been studied extensively. Skeletal muscle, liver, and the heart have primarily different metabolic changes, with few similar alterations and rare complementary alterations (alpha-tocopherol), which may illustrate the complexity of understanding exercise at the organismal level.
- Published
- 2017
- Full Text
- View/download PDF
13. Non-Targeted Metabolomics Analysis of Golden Retriever Muscular Dystrophy-Affected Muscles Reveals Alterations in Arginine and Proline Metabolism, and Elevations in Glutamic and Oleic Acid In Vivo
- Author
-
Muhammad Abdullah, Joe N. Kornegay, Aubree Honcoop, Traci L. Parry, Cynthia J. Balog-Alvarez, Sara K. O’Neal, James R. Bain, Michael J. Muehlbauer, Christopher B. Newgard, Cam Patterson, and Monte S. Willis
- Subjects
Duchenne muscular dystrophy ,golden retriever muscular dystrophy ,skeletal muscle ,metabolism ,non-targeted metabolomics ,Microbiology ,QR1-502 - Abstract
Background: Like Duchenne muscular dystrophy (DMD), the Golden Retriever Muscular Dystrophy (GRMD) dog model of DMD is characterized by muscle necrosis, progressive paralysis, and pseudohypertrophy in specific skeletal muscles. This severe GRMD phenotype includes atrophy of the biceps femoris (BF) as compared to unaffected normal dogs, while the long digital extensor (LDE), which functions to flex the tibiotarsal joint and serves as a digital extensor, undergoes the most pronounced atrophy. A recent microarray analysis of GRMD identified alterations in genes associated with lipid metabolism and energy production. Methods: We, therefore, undertook a non-targeted metabolomics analysis of the milder/earlier stage disease GRMD BF muscle versus the more severe/chronic LDE using GC-MS to identify underlying metabolic defects specific for affected GRMD skeletal muscle. Results: Untargeted metabolomics analysis of moderately-affected GRMD muscle (BF) identified eight significantly altered metabolites, including significantly decreased stearamide (0.23-fold of controls, p = 2.89 × 10−3), carnosine (0.40-fold of controls, p = 1.88 × 10−2), fumaric acid (0.40-fold of controls, p = 7.40 × 10−4), lactamide (0.33-fold of controls, p = 4.84 × 10−2), myoinositol-2-phosphate (0.45-fold of controls, p = 3.66 × 10−2), and significantly increased oleic acid (1.77-fold of controls, p = 9.27 × 10−2), glutamic acid (2.48-fold of controls, p = 2.63 × 10−2), and proline (1.73-fold of controls, p = 3.01 × 10−2). Pathway enrichment analysis identified significant enrichment for arginine/proline metabolism (p = 5.88 × 10−4, FDR 4.7 × 10−2), where alterations in L-glutamic acid, proline, and carnosine were found. Additionally, multiple Krebs cycle intermediates were significantly decreased (e.g., malic acid, fumaric acid, citric/isocitric acid, and succinic acid), suggesting that altered energy metabolism may be underlying the observed GRMD BF muscle dysfunction. In contrast, two pathways, inosine-5'-monophosphate (VIP Score 3.91) and 3-phosphoglyceric acid (VIP Score 3.08) mainly contributed to the LDE signature, with two metabolites (phosphoglyceric acid and inosine-5'-monophosphate) being significantly decreased. When the BF and LDE were compared, the most significant metabolite was phosphoric acid, which was significantly less in the GRMD BF compared to control and GRMD LDE groups. Conclusions: The identification of elevated BF oleic acid (a long-chain fatty acid) is consistent with recent microarray studies identifying altered lipid metabolism genes, while alterations in arginine and proline metabolism are consistent with recent studies identifying elevated L-arginine in DMD patient sera as a biomarker of disease. Together, these studies demonstrate muscle-specific alterations in GRMD-affected muscle, which illustrate previously unidentified metabolic changes.
- Published
- 2017
- Full Text
- View/download PDF
14. Non-Targeted Metabolomics Analysis of the Effects of Tyrosine Kinase Inhibitors Sunitinib and Erlotinib on Heart, Muscle, Liver and Serum Metabolism In Vivo
- Author
-
Brian C. Jensen, Traci L. Parry, Wei Huang, Amro Ilaiwy, James R. Bain, Michael J. Muehlbauer, Sara K. O’Neal, Cam Patterson, Gary L. Johnson, and Monte S. Willis
- Subjects
erlotinib ,sorafenib ,kinase inhibitors ,cardiotoxicity ,metabolomics ,serum ,liver ,muscle ,heart ,Microbiology ,QR1-502 - Abstract
Background: More than 90 tyrosine kinases have been implicated in the pathogenesis of malignant transformation and tumor angiogenesis. Tyrosine kinase inhibitors (TKIs) have emerged as effective therapies in treating cancer by exploiting this kinase dependency. The TKI erlotinib targets the epidermal growth factor receptor (EGFR), whereas sunitinib targets primarily vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR).TKIs that impact the function of non-malignant cells and have on- and off-target toxicities, including cardiotoxicities. Cardiotoxicity is very rare in patients treated with erlotinib, but considerably more common after sunitinib treatment. We hypothesized that the deleterious effects of TKIs on the heart were related to their impact on cardiac metabolism. Methods: Female FVB/N mice (10/group) were treated with therapeutic doses of sunitinib (40 mg/kg), erlotinib (50 mg/kg), or vehicle daily for two weeks. Echocardiographic assessment of the heart in vivo was performed at baseline and on Day 14. Heart, skeletal muscle, liver and serum were flash frozen and prepped for non-targeted GC-MS metabolomics analysis. Results: Compared to vehicle-treated controls, sunitinib-treated mice had significant decreases in systolic function, whereas erlotinib-treated mice did not. Non-targeted metabolomics analysis of heart identified significant decreases in docosahexaenoic acid (DHA), arachidonic acid (AA)/ eicosapentaenoic acid (EPA), O-phosphocolamine, and 6-hydroxynicotinic acid after sunitinib treatment. DHA was significantly decreased in skeletal muscle (quadriceps femoris), while elevated cholesterol was identified in liver and elevated ethanolamine identified in serum. In contrast, erlotinib affected only one metabolite (spermidine significantly increased). Conclusions: Mice treated with sunitinib exhibited systolic dysfunction within two weeks, with significantly lower heart and skeletal muscle levels of long chain omega-3 fatty acids docosahexaenoic acid (DHA), arachidonic acid (AA)/eicosapentaenoic acid (EPA) and increased serum O-phosphocholine phospholipid. This is the first link between sunitinib-induced cardiotoxicity and depletion of the polyunsaturated fatty acids (PUFAs) and inflammatory mediators DHA and AA/EPA in the heart. These compounds have important roles in maintaining mitochondrial function, and their loss may contribute to cardiac dysfunction.
- Published
- 2017
- Full Text
- View/download PDF
15. Correction: Cardio-Metabolic Effects of HIV Protease Inhibitors (Lopinavir/Ritonavir).
- Author
-
Kathleen M. S. E. Reyskens, Tarryn-Lee Fisher, Jonathan C. Schisler, Wendi G. O'Connor, Arlin B. Rogers, Monte S. Willis, Cynthia Planesse, Florence Boyer, Philippe Rondeau, Emmanuel Bourdon, and M. Faadiel Essop
- Subjects
Medicine ,Science - Published
- 2013
- Full Text
- View/download PDF
16. Low-Magnitude Mechanical Signals Combined with Zoledronic Acid Reduce Musculoskeletal Weakness and Adiposity in Estrogen-Deprived Mice
- Author
-
Gabriel M. Pagnotti, Trupti Trivedi, Laura E. Wright, Sutha K. John, Sreemala Murthy, Ryan R. Pattyn, Monte S. Willis, Yun She, Sukanya Suresh, William R. Thompson, Clinton T. Rubin, Khalid S. Mohammad, and Theresa A. Guise
- Subjects
Article - Abstract
Combination treatment of Low-Intensity Vibration (LIV) with zoledronic acid (ZA) was hypothesized to preserve bone mass and muscle strength while reducing adipose tissue accrual associated with complete estrogen (E2)-deprivation in young and skeletally mature mice. Complete E2-deprivation (surgical-ovariectomy (OVX) and daily injection of aromatase inhibitor (AI) letrozole) were performed on 8-week-old C57BL/6 female mice for 4 weeks following commencement of LIV administration or control (no LIV), for 28 weeks. Additionally, 16-week-old C57BL/6 female E2-deprived mice were administered ±LIV twice daily and supplemented with ±ZA (2.5 ng/kg/week). By week 28, lean tissue mass quantified by dual-energy X-ray absorptiometry was increased in younger OVX/AI+LIV(y) mice, with increased myofiber cross-sectional area of quadratus femorii. Grip strength was greater in OVX/AI+LIV(y) mice than OVX/AI(y) mice. Fat mass remained lower in OVX/AI+LIV(y) mice throughout the experiment compared with OVX/AI(y) mice. OVX/AI+LIV(y) mice exhibited increased glucose tolerance and reduced leptin and free fatty acids than OVX/AI(y) mice. Trabecular bone volume fraction and connectivity density increased in the vertebrae of OVX/AI+LIV(y) mice compared to OVX/AI(y) mice; however, this effect was attenuated in the older cohort of E2-deprived mice, specifically in OVX/AI+ZA mice, requiring combined LIV with ZA to increase trabecular bone volume and strength. Similar improvements in cortical bone thickness and cross-sectional area of the femoral mid-diaphysis were observed in OVX/AI+LIV+ZA mice, resulting in greater fracture resistance. Our findings demonstrate that the combination of mechanical signals in the form of LIV and anti-resorptive therapy via ZA improve vertebral trabecular bone and femoral cortical bone, increase lean mass, and reduce adiposity in mice undergoing complete E2-deprivation.One Sentence Summary:Low-magnitude mechanical signals with zoledronic acid suppressed bone and muscle loss and adiposity in mice undergoing complete estrogen deprivation.Translational RelevancePostmenopausal patients with estrogen receptor-positive breast cancer treated with aromatase inhibitors to reduce tumor progression experience deleterious effects to bone and muscle subsequently develop muscle weakness, bone fragility, and adipose tissue accrual. Bisphosphonates (i.e., zoledronic acid) prescribed to inhibit osteoclast-mediated bone resorption are effective in preventing bone loss but may not address the non-skeletal effects of muscle weakness and fat accumulation that contribute to patient morbidity. Mechanical signals, typically delivered to the musculoskeletal system during exercise/physical activity, are integral for maintaining bone and muscle health; however, patients undergoing treatments for breast cancer often experience decreased physical activity which further accelerates musculoskeletal degeneration. Low-magnitude mechanical signals, in the form of low-intensity vibrations, generate dynamic loading forces similar to those derived from skeletal muscle contractility. As an adjuvant to existing treatment strategies, low-intensity vibrations may preserve or rescue diminished bone and muscle degraded by breast cancer treatment.
- Published
- 2023
17. Correction: The Role of Bone Muscle Ring Finger-1 (MuRF1), MuRF2, MuRF3, and Atrogin-1 on Microarchitecture In Vivo
- Author
-
Vidyani Suryadevara, Connor J. Krehbial, Danielle Halsey, and Monte S. Willis
- Subjects
Biophysics ,Cell Biology ,General Medicine ,Biochemistry - Published
- 2022
18. The Role of Muscle Ring Finger-1 (MuRF1), MuRF2, MuRF3, and Atrogin-1 on Bone Microarchitecture In Vivo
- Author
-
Vidyani Suryadevara, Connor J. Krehbial, Danielle Halsey, and Monte S. Willis
- Subjects
Biophysics ,Cell Biology ,General Medicine ,Biochemistry - Published
- 2022
19. Chemokine-Based Therapeutics for the Treatment of Inflammatory and Fibrotic Convergent Pathways in COVID-19
- Author
-
Dana R. Julian, Megan A. Kazakoff, Akhil Patel, Jesse Jaynes, Monte S. Willis, and Cecelia C. Yates
- Subjects
Cancer Research ,CXCL10 ,Wound Healing and Tissue Repair (Cc Yates, Section Editor) ,COVID-19 ,Cell Biology ,Chemokines ,Fibrosis ,Molecular Biology ,Pathology and Forensic Medicine - Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by the SARS-CoV-2 betacoronavirus and has taken over 761,426 American lives as of the date of publication and will likely result in long-term, if not permanent, tissue damage for countless patients. COVID-19 presents with diverse and multisystemic pathologic processes, including a hyperinflammatory response, acute respiratory distress syndrome (ARDS), vascular injury, microangiopathy, tissue fibrosis, angiogenesis, and widespread thrombosis across multiple organs, including the lungs, heart, kidney, liver, and brain. C-X-C chemokines contribute to these pathologies by attracting inflammatory mediators, the disruption of endothelial cell integrity and function, and the initiation and propagation of the cytokine storm. Among these, CXCL10 is recognized as a critical contributor to the hyperinflammatory state and poor prognosis in COVID-19. CXCL10 is also known to regulate growth factor-induced fibrosis, and recent evidence suggests the CXCL10-CXCR3 signaling system may be vital in targeting convergent pro-inflammatory and pro-fibrotic pathways. This review will explore the mechanistic role of CXCL10 and related chemokines in fibrotic complications associated with COVID-19 and the potential of CXCL10-targeted therapeutics for early intervention and long-term treatment of COVID-19-induced fibrosis.
- Published
- 2021
20. ACVR2B antagonism as a countermeasure to multi‐organ perturbations in metastatic colorectal cancer cachexia
- Author
-
Fabrizio Pin, Andrea Bonetto, Leah J. Novinger, Monte S. Willis, Joshua R. Huot, Ashok Narasimhan, Austin S Keith, and Teresa A. Zimmers
- Subjects
Male ,0301 basic medicine ,Cardiac function curve ,medicine.medical_specialty ,Cachexia ,Skeletal muscle ,Adipose tissue ,Liver metastases ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Orthopedics and Sports Medicine ,Bone ,Muscle, Skeletal ,Wasting ,Ejection fraction ,business.industry ,Liver Neoplasms ,Cardiac muscle ,Cancer ,Heart ,Original Articles ,medicine.disease ,Colorectal cancer ,Disease Models, Animal ,Activin signalling ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,030220 oncology & carcinogenesis ,Original Article ,medicine.symptom ,Colorectal Neoplasms ,business - Abstract
Background Advanced colorectal cancer (CRC) is often accompanied by the development of liver metastases, as well as cachexia, a multi‐organ co‐morbidity primarily affecting skeletal (SKM) and cardiac muscles. Activin receptor type 2B (ACVR2B) signalling is known to cause SKM wasting, and its inhibition restores SKM mass and prolongs survival in cancer. Using a recently generated mouse model, here we tested whether ACVR2B blockade could preserve multiple organs, including skeletal and cardiac muscle, in the presence of metastatic CRC. Methods NSG male mice (8 weeks old) were injected intrasplenically with HCT116 human CRC cells (mHCT116), while sham‐operated animals received saline (n = 5–10 per group). Sham and tumour‐bearing mice received weekly injections of ACVR2B/Fc, a synthetic peptide inhibitor of ACVR2B. Results mHCT116 hosts displayed losses in fat mass ( − 79%, P
- Published
- 2020
21. The Unraveling
- Author
-
Monte S. Willis, Michael Klüppel, Vidyani Suryadevara, and Federica del Monte
- Subjects
0301 basic medicine ,Systemic disease ,Bone density ,business.industry ,Diastolic heart failure ,Muscle weakness ,Disease ,medicine.disease ,Bioinformatics ,Muscle atrophy ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Heart failure ,medicine ,medicine.symptom ,Alzheimer's disease ,business ,030217 neurology & neurosurgery - Abstract
Alzheimer disease (AD) is characterized by deterioration of cognitive capabilities with an estimated 44 million individuals worldwide living with it. Beyond memory deficits, the most common AD co-morbidities include swallowing defects (muscle), fractures (bone, muscle), and heart failure. The underlying causes of these co-morbidities and their role in AD pathophysiology are currently unknown. This review is the first to summarize the emerging picture of the cardiac and musculoskeletal deficits in human AD. We present the involvement of the heart, characterized by diastolic heart failure, the presence of amyloid deposits, and electrophysiological changes, compared with age-matched control subjects. The characteristic musculoskeletal defects in AD come from recent clinical studies and include potential underlying mechanisms (bone) in animal models. These studies detail a primary muscle weakness (without a loss of muscle mass) in patients with mild cognitive impairment, with progression of cognitive impairment to AD associating with ongoing muscle weakness and the onset of muscle atrophy. We conclude by reviewing the loss of bone density in patients with AD, paralleling the increase in fracture and fall risk in specific populations. These studies paint AD as a systemic disease in broad strokes, which may help elucidate AD pathophysiology and to allow for new ways of thinking about therapeutic interventions, diagnostic biomarkers, and the pathogenesis of this multidisciplinary disease.
- Published
- 2020
22. The Role of Muscle Ring Finger-1 (MuRF1), MuRF2, MuRF3, and Atrogin-1 on Bone Microarchitecture In Vivo
- Author
-
Vidyani, Suryadevara, Connor J, Krehbial, Danielle, Halsey, and Monte S, Willis
- Subjects
Male ,Tripartite Motif Proteins ,Mice ,SKP Cullin F-Box Protein Ligases ,Ubiquitin ,Muscles ,Ubiquitin-Protein Ligases ,Animals ,Muscle Proteins ,Female ,Muscle, Skeletal - Abstract
Ubiquitin proteasome system was found to contribute to bone loss by regulating bone turnover and metabolism, by modulating osteoblast differentiation and bone formation as well as formation of osteoclasts that contribute to bone resorption. Muscle Ring Finger (MuRF) are novel ubiquitin ligases, which are muscle specific and have not been much implicated in the bone but have been implicated in several human diseases including heart failure and skeletal muscle atrophy. This study is aimed at understanding the role of MuRF1, MuRF2, MuRF3 and Atrogin which are distinct MuRF family proteins in bone homeostasis. Wildtype, heterozygous and homozygous mice of each of the isoforms were used and the bone microarchitecture and mechanical properties were assessed using microCT and biomechanics. MuRF1 depletion was found to alter cortical properties in both males and females, but only trabecular spacing in the females. MuRF2 depletion let to no changes in the cortical and trabecular properties but change in the strain to yield in the females. Depletion of MuRF3 led to decrease in the cortical properties in the females and increase in the trabecular properties in the males. Atrogin depletion was found to reduce cortical properties in both males and females, whereas some trabecular properties were found to be reduced in the females. Each muscle-specific ligase was found to alter the bone structure and mechanical properties in a distinct a sex-dependent manner.
- Published
- 2021
23. Muscle RING finger-1 is required to prevent age-related cardiac hypertrophy and interstitial remodelling
- Author
-
Valentina Prando, Traci L. Parry, Vidyani Suryadevara, Arianna Scalco, Nicola Pianca, Nicola Moro, Paola Pesce, Wei Tang, Hong Ma, Li Qian, Jiandong Liu, Maddalena Tessari, Raffaele Coppini, Elisabetta Cerbai, Cam Patterson, Cristina Basso, Giuseppe Faggian, Paolo Bonaldo, Marco Sandri, Marco Mongillo, Monte S. Willis, and Tania Zaglia
- Subjects
Pharmacology ,Physiology ,Molecular Medicine - Published
- 2022
24. F-box protein-32 down-regulates small-conductance calcium-activated potassium channel 2 in diabetic mouse atria
- Author
-
Tian You Ling, Hon Chi Lee, Tong Lu, Xiao Li Wang, Monte S. Willis, Fu Yi, Li Qun Wu, John P. Adelman, Xiaojing Sun, and Win Kuang Shen
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Vascular smooth muscle ,Small-Conductance Calcium-Activated Potassium Channels ,Down-Regulation ,Muscle Proteins ,Protein degradation ,Biochemistry ,Streptozocin ,Diabetes Mellitus, Experimental ,SK channel ,Mice ,03 medical and health sciences ,Tumor Cells, Cultured ,Animals ,Molecular Biology ,Gene knockdown ,SKP Cullin F-Box Protein Ligases ,030102 biochemistry & molecular biology ,biology ,Ubiquitin ,Chemistry ,HEK 293 cells ,Molecular Bases of Disease ,Cell Biology ,Calcium-activated potassium channel ,Potassium channel ,Cell biology ,Ubiquitin ligase ,030104 developmental biology ,biology.protein - Abstract
Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation, but the underlying ionic mechanism for this association remains unclear. We recently reported that expression of the small-conductance calcium-activated potassium channel 2 (SK2, encoded by KCCN2) in atria from diabetic mice is significantly down-regulated, resulting in reduced SK currents in atrial myocytes from these mice. We also reported that the level of SK2 mRNA expression is not reduced in DM atria but that the ubiquitin-proteasome system (UPS), a major mechanism of intracellular protein degradation, is activated in vascular smooth muscle cells in DM. This suggests a possible role of the UPS in reduced SK currents. To test this possibility, we examined the role of the UPS in atrial SK2 down-regulation in DM. We found that a muscle-specific E3 ligase, F-box protein 32 (FBXO-32, also called atrogin-1), was significantly up-regulated in diabetic mouse atria. Enhanced FBXO-32 expression in atrial cells significantly reduced SK2 protein expression, and siRNA-mediated FBXO-32 knockdown increased SK2 protein expression. Furthermore, co-transfection of SK2 with FBXO-32 complementary DNA in HEK293 cells significantly reduced SK2 expression, whereas co-transfection with atrogin-1ΔF complementary DNA (a nonfunctional FBXO-32 variant in which the F-box domain is deleted) did not have any effects on SK2. These results indicate that FBXO-32 contributes to SK2 down-regulation and that the F-box domain is essential for FBXO-32 function. In conclusion, DM-induced SK2 channel down-regulation appears to be due to an FBXO-32-dependent increase in UPS-mediated SK2 protein degradation.
- Published
- 2019
25. Image-Based Methods for Phase Estimation, Gating, and Temporal Superresolution of Cardiac Ultrasound
- Author
-
Samuel Gerber, Marc Niethammer, Deepak Roy Chittajallu, Roland Kwitt, Stephen R. Aylward, Monte S. Willis, Ryan C. Gessner, Tomasz J. Czernuszewicz, and Matt McCormick
- Subjects
Cardiac function curve ,Computer science ,Video Recording ,Biomedical Engineering ,030204 cardiovascular system & hematology ,010502 geochemistry & geophysics ,01 natural sciences ,Article ,Mice ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Ultrasound ,Image Processing, Computer-Assisted ,medicine ,Animals ,Temporal Super-resolution ,Image resolution ,0105 earth and related environmental sciences ,medicine.diagnostic_test ,business.industry ,Phase estimation ,Frame (networking) ,Heart ,Pattern recognition ,Cardiorespiratory fitness ,Echocardiography ,Temporal resolution ,symbols ,Hilbert transform ,Artificial intelligence ,business ,Cardiac ,Gating ,Electrocardiography ,Algorithms - Abstract
Objective: Ultrasound is an effective tool for rapid noninvasive assessment of cardiac structure and function. Determining the cardiorespiratory phases of each frame in the ultrasound video and capturing the cardiac function at a much higher temporal resolution are essential in many applications. Fulfilling these requirements is particularly challenging in preclinical studies involving small animals with high cardiorespiratory rates, requiring cumbersome and expensive specialized hardware. Methods: We present a novel method for the retrospective estimation of cardiorespiratory phases directly from the ultrasound videos. It transforms the videos into a univariate time series preserving the evidence of periodic cardiorespiratory motion, decouples the signatures of cardiorespiratory motion with a trend extraction technique, and estimates the cardiorespiratory phases using a Hilbert transform approach. We also present a robust nonparametric regression technique for respiratory gating and a novel kernel-regression model for reconstructing images at any cardiac phase facilitating temporal superresolution. Results: We validated our methods using two-dimensional echocardiography videos and electrocardiogram (ECG) recordings of six mice. Our cardiac phase estimation method provides accurate phase estimates with a mean-phase-error range of 3%–6% against ECG derived phase and outperforms three previously published methods in locating ECGs R-wave peak frames with a mean-frame-error range of 0.73–1.36. Our kernel-regression model accurately reconstructs images at any cardiac phase with a mean-normalized-correlation range of 0.81–0.85 over 50 leave-one-out-cross-validation rounds. Conclusion and significance: Our methods can enable tracking of cardiorespiratory phases without additional hardware and reconstruction of respiration-free single cardiac-cycle videos at a much higher temporal resolution.
- Published
- 2019
26. Muscle ring finger-1 is required to prevent age-related cardiac hypertrophy and interstitial remodelling
- Author
-
Cristina Basso, W Tang, P Braghetta, V Prando, Tania Zaglia, Paola Pesce, Marco Sandri, Paolo Bonaldo, Giuseppe Faggian, Monte S. Willis, Traci L. Parry, Arianna Scalco, H Ma, and Marco Mongillo
- Subjects
medicine.medical_specialty ,Muscle ring finger ,business.industry ,Internal medicine ,Age related ,Cardiac hypertrophy ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
Purpose The Ubiquitin Proteasome System (UPS) is a selective degradation system mediating the removal of intracellular unfolded/misfolded proteins and is essential for cardiomyocyte (CM) health. Substrate specificity and ubiquitination rate are mediated by E3 ubiquitin-ligases, such as Atrogin1 and MuRF1, which are specifically expressed in muscle cells. Perturbation of protein quality control causes aggregation of misfolded proteins, leading to CM proteotoxicity. UPS dysfunction occurs in ageing, a risk factor for cardiac hypertrophy and HF. We recently demonstrated that Atrogin-1 is essential to maintain CM health, during ageing. Whether MuRF1 plays similar roles in heart adaptation to ageing is still unexplored, and different studies have yielded contrasting results. Methods To assess the role of MuRF1 in heart homeostasis, we combined echocardiography, histology, IF, TUNEL assay and EM on heart sections from MuRF1 knock-out (KO) mice, and littermate controls, at 3, 10 and 24 mo. RTqPCR and WB assessed markers of UPS and extracellular matrix. Langendorff procedure was used to separate CMs from cardiac fibroblasts. Molecular and IF analyses were performed in heart samples from patients affected by aortic stenosis. Results MuRF1 ablation leads to cardiac hypertrophy, progressing during ageing (LV CM areas: 3 mo., KO: 329.14±10.66 vs Ctrl: 296.25±5.43; 10 mo., KO: 399.73±7.64 vs Ctrl: 247.49±3.67; 24 mo., KO: 418.89±11.10 vs. Ctrl: 209.93±4.48, in μm2). The hypertrophic remodeling was accompanied by diastolic dysfunction in the adulthood and, during ageing, also by systolic dysfunction (EF, 24 mo., KO: 29.55±8.82 vs Ctrl: 51.23±6.56, in %). Loss of MuRF1 causes increased interstitial collagen -I and -VI deposition, even before the onset of contractile dysfunction, followed by activation of Matrix MetalloProteinases (MMPs), suggesting that such alterations may be responsible for decreased cardiac performance. Interestingly, collagen established rings enveloping MuRF1 KO CMs and such fibrotic remodeling was not accompanied by increased CM apoptosis, nor myofibroblast activation. Such peculiar remodelling, called peri-endomysial fibrosis, was detected in hearts from patients with aortic stenosis, a condition in which MuRF1 levels decrease. Our data supports that MuRF1 has a role in CM-dependent regulation of the extracellular matrix (ECM) dynamics. Consistently, MuRF1 downregulation in normal cultured CMs demonstrated that such ubiquitin ligase impacts on signaling pathways involved in the control of the ECM homeostasis. In addition, fibroblasts treated with culture medium conditioned by MuRF1 KO CM display increased collagen and MMP expression. Conclusions We identifieded a novel role of MuRF1 in the control of CM proteostasis, and unveiled that in addition to cardiac fibroblasts, CM may directly regulate ECM dynamics, indicating that the correct function of MuRF1 is essential for heart adaptation to aging. Funding Acknowledgement Type of funding source: Public grant(s) – National budget only. Main funding source(s): University of Padova
- Published
- 2020
27. 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
28. The Access Technology Program of the Indiana Clinical Translational Sciences Institute (CTSI): A model to facilitate access to cutting-edge technologies across a state
- Author
-
Jenna York, Joel A. Ybe, Jeanette N. McClintick, Christie M. Orschell, Lilith Reeves, Kenneth Cornetta, Joe D. Hunt, Monte S. Willis, Thomas G. Sors, Jill L. Reiter, Julie Driscol, Todd C. Skaar, Melanie E. DeFord, Christine Drury, Anantha Shekhar, and Rob Orr
- Subjects
0301 basic medicine ,CTSI ,Engineering ,030102 biochemistry & molecular biology ,Access technology ,business.industry ,pilot funding ,Best practice ,Research Methods and Technology ,Translational research ,General Medicine ,core facilities ,03 medical and health sciences ,Engineering management ,030104 developmental biology ,technology ,Mandate ,State (computer science) ,Translational science ,business ,Research Article - Abstract
Introduction: Access to cutting-edge technologies is essential for investigators to advance translational research. The Indiana Clinical and Translational Sciences Institute (CTSI) spans three major and preeminent universities, four large academic campuses across the state of Indiana, and is mandate to provide best practices to a whole state. Methods: To address the need to facilitate the availability of innovative technologies to its investigators, the Indiana CTSI implemented the Access Technology Program (ATP). The activities of the ATP, or any program of the Indiana CTSI, are challenged to connect technologies and investigators on the multiple Indiana CTSI campuses by the geographical distances between campuses (1–4 hr driving time). Results: Herein, we describe the initiatives developed by the ATP to increase the availability of state-of-the-art technologies to its investigators on all Indiana CTSI campuses, and the methods developed by the ATP to bridge the distance between campuses, technologies, and investigators for the advancement of clinical translational research. Conclusions: The methods and practices described in this publication may inform other approaches to enhance translational research, dissemination, and usage of innovative technologies by translational investigators, especially when distance or multi-campus cultural differences are factors to efficient application.
- Published
- 2020
29. Abstract MP101: Functional Maintenance of the Sarcomere Requires Bag3-dependent Autophagy
- Author
-
Arthur M. Feldman, Thomas G Martin, Monte S. Willis, and Jonathan A. Kirk
- Subjects
Physiology ,Autophagy ,Biology ,Cardiology and Cardiovascular Medicine ,BAG3 ,Sarcomere ,Cell biology - Abstract
Bcl2-associated athanogene-3 (BAG3) is a pro-autophagy co-chaperone highly expressed in the heart. Clinical studies show BAG3 haploinsufficiency and mutations are associated with heart failure (HF). However, these studies are largely observational and fail to go beyond the observed phenotype and study mechanism. One study in neonatal myocytes suggests a role for BAG3 in structural maintenance of the sarcomere. However, the structural and functional significance of BAG3 in adult myocytes is not known. We found that myofilament BAG3 expression decreases in human heart failure and is associated with impaired myofilament force-generating capacity (F max ). To assess whether rescuing BAG3 levels could restore function, we used a mouse model of HF and treated with BAG3 gene therapy via AAV9. Myofilament function was assessed in skinned cardiomyocytes by force-calcium relationship. HF mice experienced a reduction in F max , but this was fully restored to sham levels by BAG3 gene therapy.To explore mechanism, we used mass spectrometry to identify the BAG3-interactome at the myofilament and found heat shock proteins (HSP) 70 and B8 among the top hits. Immunofluorescence further showed that HSP70, B8, and BAG3 each localized to the sarcomere z-disk. This BAG3-Hsp complex had previously been described to promote ubiquitin-dependent autophagy in skeletal muscle. Notably, in both human HF samples and in the mouse HF model, myofilament ubiquitin levels increased significantly. However, BAG3 gene therapy in HF reduces ubiquitin levels and restores autophagy flux. This suggests that BAG3 serves a role in proteostasis for the sarcomere, which may explain the functional effect of BAG3 gene therapy.To further explore the impact of the BAG3/HSP complex on myofilament function, we used a mouse model with the P209L BAG3 mutation, which had previously been described to disrupt client processing by the complex. We found cardiomyocytes from P209L mice had significantly reduced F max and elevated myofilament ubiquitin levels, suggesting BAG3-dependent autophagy is required to maintain function. Together, our data identify a functional role for BAG3 at the sarcomere and indicate BAG3-mediated autophagy is an important mechanism for maintaining myofilament proteostasis.
- Published
- 2020
30. Cardiomyocyte contractile impairment in heart failure results from reduced BAG3-mediated sarcomeric protein turnover
- Author
-
Arthur M. Feldman, Edith Perez, Monte S. Willis, Praveen K Dubey, Valerie D. Myers, Christine S. Moravec, Shubham Dubey, Jonathan A. Kirk, and Thomas G Martin
- Subjects
0301 basic medicine ,Male ,Proteomics ,Myofilament ,General Physics and Astronomy ,Muscle Proteins ,030204 cardiovascular system & hematology ,Sarcomere ,Rats, Sprague-Dawley ,Mice ,0302 clinical medicine ,Ubiquitin ,Myocytes, Cardiac ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,Middle Aged ,Recombinant Proteins ,Cell biology ,medicine.anatomical_structure ,Mechanisms of disease ,Female ,Adult ,Sarcomeres ,Science ,Heart failure ,BAG3 ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,medicine ,Autophagy ,Animals ,Humans ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Aged ,Protein turnover ,Skeletal muscle ,General Chemistry ,Genetic Therapy ,medicine.disease ,Myocardial Contraction ,Rats ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Proteostasis ,biology.protein ,Apoptosis Regulatory Proteins - Abstract
The association between reduced myofilament force-generating capacity (Fmax) and heart failure (HF) is clear, however the underlying molecular mechanisms are poorly understood. Here, we show impaired Fmax arises from reduced BAG3-mediated sarcomere turnover. Myofilament BAG3 expression decreases in human HF and positively correlates with Fmax. We confirm this relationship using BAG3 haploinsufficient mice, which display reduced Fmax and increased myofilament ubiquitination, suggesting impaired protein turnover. We show cardiac BAG3 operates via chaperone-assisted selective autophagy (CASA), conserved from skeletal muscle, and confirm sarcomeric CASA complex localization is BAG3/proteotoxic stress-dependent. Using mass spectrometry, we characterize the myofilament CASA interactome in the human heart and identify eight clients of BAG3-mediated turnover. To determine if increasing BAG3 expression in HF can restore sarcomere proteostasis/Fmax, HF mice were treated with rAAV9-BAG3. Gene therapy fully rescued Fmax and CASA protein turnover after four weeks. Our findings indicate BAG3-mediated sarcomere turnover is fundamental for myofilament functional maintenance., Decreased expression of BAG3 in the heart is associated with contractile dysfunction and heart failure. Here the authors show that this is due to decreased BAG3-dependent sarcomere protein turnover, which impairs mechanical function, and that sarcomere force-generating capacity is restored with BAG3 gene therapy.
- Published
- 2020
31. The Unraveling: Cardiac and Musculoskeletal Defects and Their Role in Common Alzheimer Disease Morbidity and Mortality
- Author
-
Vidyani, Suryadevara, Michael, Klüppel, Federica Del, Monte, and Monte S, Willis
- Subjects
Muscle Weakness ,Alzheimer Disease ,Disease Progression ,Humans ,Cognitive Dysfunction ,Heart ,Muscle, Skeletal - Abstract
Alzheimer disease (AD) is characterized by deterioration of cognitive capabilities with an estimated 44 million individuals worldwide living with it. Beyond memory deficits, the most common AD co-morbidities include swallowing defects (muscle), fractures (bone, muscle), and heart failure. The underlying causes of these co-morbidities and their role in AD pathophysiology are currently unknown. This review is the first to summarize the emerging picture of the cardiac and musculoskeletal deficits in human AD. We present the involvement of the heart, characterized by diastolic heart failure, the presence of amyloid deposits, and electrophysiological changes, compared with age-matched control subjects. The characteristic musculoskeletal defects in AD come from recent clinical studies and include potential underlying mechanisms (bone) in animal models. These studies detail a primary muscle weakness (without a loss of muscle mass) in patients with mild cognitive impairment, with progression of cognitive impairment to AD associating with ongoing muscle weakness and the onset of muscle atrophy. We conclude by reviewing the loss of bone density in patients with AD, paralleling the increase in fracture and fall risk in specific populations. These studies paint AD as a systemic disease in broad strokes, which may help elucidate AD pathophysiology and to allow for new ways of thinking about therapeutic interventions, diagnostic biomarkers, and the pathogenesis of this multidisciplinary disease.
- Published
- 2020
32. Cardiomyocyte-specific ACSL1 Deficiency Prevents Cardiac Lipotoxicity and Alleviates Heart Dysfunction in the ob/ob Model of Obesity
- Author
-
Florencia Pascual, Liyang Zhao, Monte S. Willis, Trisha J. Grevengoed, and Rosalind A. Coleman
- Subjects
0303 health sciences ,medicine.medical_specialty ,Gene knockdown ,business.industry ,Cardiomyopathy ,Diastole ,Context (language use) ,030204 cardiovascular system & hematology ,medicine.disease ,Muscle hypertrophy ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Lipotoxicity ,Internal medicine ,Gene expression ,medicine ,business ,Tamoxifen ,030304 developmental biology ,medicine.drug - Abstract
Cardiac lipotoxicity is associated with structural remodeling and functional changes that are features of obesity-related cardiomyopathy. Both high fat diet and the ob/ob mutation lead to increased fatty acid (FA) uptake, elevated triacylglycerol (TAG) content, hypertrophy, and systolic and diastolic dysfunction in murine hearts. Cardiomyocyte-specific long-chain acyl-CoA synthetase 1 (ACSL1) deficiency (Acsl1H-/-) results in a 90% reduction in FA activation, suggesting that Acsl1 ablation might alleviate obesity-associated myocardial dysfunction. Double knockout ob-Acsl1H-/- and ob-Acsl1flox/flox control mice were treated with tamoxifen at 20 weeks of age; heart function, TAG content, and relevant gene expression were assessed immediately before and 2 and 5 weeks after treatment. Heart weights initially increased in lean and obese Acsl1H-/- mice, but normalized in ob-Acsl1H-/- mice by 5 weeks. Ventricular TAG content was decreased by 51% and 61% in ob-Acsl1H-/- mice 2 and 5 weeks after Acsl1 knockout induction, respectively. Moreover, ACSL1 knockout resulted in increased survival of ob/ob mice, suggesting that lack of ACSL1 protected obese hearts subjected to stress. Our results indicate that partial knockdown of cardiac ACSL1 is sufficient to reverse cardiac TAG accumulation and to ameliorate heart dysfunction even in the context of established obesity-related cardiomyopathy.
- Published
- 2020
- Full Text
- View/download PDF
33. New insights into immunomodulation via overexpressing lipoic acid synthase as a therapeutic potential to reduce atherosclerosis
- Author
-
Roberto Mota, Stephen W. Simington, Joseph M. DeSimone, Darcy Holley, Xianwen Yi, Jun Nakamura, Zhenquan Jia, Nobuyo Maeda, Monte S. Willis, Shaomin Tian, Sylvia Hiller, J. Christopher Luft, and Scott J. Bultman
- Subjects
0301 basic medicine ,Male ,Antioxidant ,Apolipoprotein B ,Physiology ,Mice, Knockout, ApoE ,medicine.medical_treatment ,T cell ,Aortic Diseases ,Endogeny ,030204 cardiovascular system & hematology ,Pharmacology ,T-Lymphocytes, Regulatory ,Article ,Lesion ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Gene ,Aorta ,Autoantibodies ,biology ,Chemistry ,Autoantibody ,Atherosclerosis ,Plaque, Atherosclerotic ,Lipoproteins, LDL ,Mice, Inbred C57BL ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,Enzyme Induction ,Sulfurtransferases ,biology.protein ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Oxidation-Reduction - Abstract
Atherosclerosis is a systemic chronic inflammatory disease. Many antioxidants including alpha-lipoic acid (LA), a product of lipoic acid synthase (Lias), have proven to be effective for treatment of this disease. However, the question remains whether LA regulates the immune response as a protective mechanism against atherosclerosis. We initially investigated whether enhanced endogenous antioxidant can retard the development of atherosclerosis via immunomodulation. To explore the impact of enhanced endogenous antioxidant on the retardation of atherosclerosis via immune regulation, our laboratory has recently created a double mutant mouse model, using apolipoprotein E-deficient (Apoe(−/−)) mice crossbred with mice overexpressing lipoic acid synthase gene (Lias(H/H)), designated as Lias(H/H)Apoe(−/−) mice. Their littermates, Lias(+/+)Apoe(−/−) mice, served as a control. Distinct redox environments between the two strains of mice have been established and they can be used to facilitate identification of antioxidant targets in the immune response. At 6 months of age, Lias(H/H)Apoe(−/−) mice had profoundly decreased atherosclerotic lesion size in the aortic sinus compared to their Lias(+/+)Apoe(−/−) littermates, accompanied by significantly enhanced numbers of regulatory T cells (Tregs) and anti-oxidized LDL autoantibody in the vascular system, and reduced T cell infiltrates in aortic walls. Our results represent a novel exploration into an environment with increased endogenous antioxidant and its ability to alleviate atherosclerosis, likely through regulation of the immune response. These outcomes shed light on a new therapeutic strategy using antioxidants to lessen atherosclerosis.
- Published
- 2020
34. Tumor necrosis factor receptor-associated factor 6 as a nuclear factor kappa B-modulating therapeutic target in cardiovascular diseases: at the heart of it all
- Author
-
Muhammad Abdullah, Monte S. Willis, and Jessica M. Berthiaume
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Myocarditis ,Heart disease ,Cardiomyopathy ,Inflammation ,Disease ,Bioinformatics ,Article ,03 medical and health sciences ,Physiology (medical) ,medicine ,Animals ,Humans ,TNF Receptor-Associated Factor 6 ,Ubiquitin ,business.industry ,Biochemistry (medical) ,NF-kappa B ,Public Health, Environmental and Occupational Health ,General Medicine ,medicine.disease ,Pathophysiology ,030104 developmental biology ,Cardiovascular Diseases ,Heart failure ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Signal Transduction - Abstract
Inflammatory and immune signaling has been documented as a root cause of many cardiovascular pathologies. In this review, we explore the emerging role of tumor necrosis factor receptor-associated factor 6 (TRAF6)-nuclear factor kappa B (NF-κB) signaling axis in atherosclerosis, ischemic heart disease, pathologic cardiac hypertrophy or heart failure, myocarditis, and sepsis-induced cardiomyopathy. We discuss the current understanding of cardiac inflammation in heart disease, present the TRAF6 signaling axis in the heart, then summarize what is known about TRAF6 in pathophysiology of heart disease including proof-of-concept studies that identify the utility of blocking TRAF6 to attenuate cardiac dysfunction, which suggests that TRAF6 is a novel, druggable target in treating cardiovascular disease incurred by inflammatory processes.
- Published
- 2018
35. Genome-wide association study of homocysteine in African Americans from the Jackson Heart Study, the Multi-Ethnic Study of Atherosclerosis, and the Coronary Artery Risk in Young Adults study
- Author
-
Laura M. Raffield, Nels C. Olson, Mary Cushman, James G. Wilson, Ethan M. Lange, Nathan Pankratz, Peter Durda, Leslie A. Lange, Qing Duan, Myron D. Gross, Jin Li, Christina L. Wassel, Russell P. Tracy, Yun Li, Stephen S. Rich, Monte S. Willis, Brendan J. Keating, Jaclyn Ellis, and Alexander P. Reiner
- Subjects
Adult ,Male ,0301 basic medicine ,Genotype ,Homocysteine ,Quantitative Trait Loci ,Physiology ,Locus (genetics) ,Genome-wide association study ,Coronary Artery Disease ,Disease ,Quantitative trait locus ,Polymorphism, Single Nucleotide ,Article ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,Mississippi ,Quantitative Trait, Heritable ,Genetics ,Humans ,Medicine ,Genetic Predisposition to Disease ,Longitudinal Studies ,Young adult ,Allele ,Alleles ,Genetics (clinical) ,business.industry ,Middle Aged ,Atherosclerosis ,MMACHC ,3. Good health ,Black or African American ,030104 developmental biology ,chemistry ,Population Surveillance ,Female ,business ,Genome-Wide Association Study - Abstract
Homocysteine (Hcy) is a heritable biomarker for CVD, peripheral artery disease, stroke, and dementia. Little is known about genetic associations with Hcy in individuals of African ancestry. We performed a genome-wide association study for Hcy in 4927 AAs from the Jackson Heart Study (JHS), the Multi-Ethnic Study of Atherosclerosis (MESA), and the Coronary Artery Risk in Young Adults (CARDIA) study. Analyses were stratified by sex and results were meta-analyzed within and across sex. In the sex-combined meta-analysis, we observed genome-wide significant evidence (p
- Published
- 2018
36. Effects of the kinase inhibitor sorafenib on heart, muscle, liver and plasma metabolism in vivo using non-targeted metabolomics analysis
- Author
-
Christopher B. Newgard, Wei Huang, Amro Ilaiwy, Cam Patterson, Ju Youn Beak, Michael J. Muehlbauer, Traci L. Parry, Monte S. Willis, Brian C. Jensen, Gary L. Johnson, and James R. Bain
- Subjects
0301 basic medicine ,Pharmacology ,Sorafenib ,medicine.medical_specialty ,Cardiotoxicity ,Taurine ,Kinase ,Skeletal muscle ,Hypotaurine ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,In vivo ,Internal medicine ,medicine ,Kinome ,medicine.drug - Abstract
Background and purpose The human kinome consists of roughly 500 kinases, including 150 that have been proposed as therapeutic targets. Protein kinases regulate an array of signalling pathways that control metabolism, cell cycle progression, cell death, differentiation and survival. It is not surprising, then, that new kinase inhibitors developed to treat cancer, including sorafenib, also exhibit cardiotoxicity. We hypothesized that sorafenib cardiotoxicity is related to its deleterious effects on specific cardiac metabolic pathways given the critical roles of protein kinases in cardiac metabolism. Experimental approach FVB/N mice (10 per group) were challenged with sorafenib or vehicle control daily for 2 weeks. Echocardiographic assessment of the heart identified systolic dysfunction consistent with cardiotoxicity in sorafenib-treated mice compared to vehicle-treated controls. Heart, skeletal muscle, liver and plasma were flash frozen and prepped for non-targeted GC-MS metabolomics analysis. Key results Compared to vehicle-treated controls, sorafenib-treated hearts exhibited significant alterations in 11 metabolites, including markedly altered taurine/hypotaurine metabolism (25-fold enrichment), identified by pathway enrichment analysis. Conclusions and implications These studies identified alterations in taurine/hypotaurine metabolism in the hearts and skeletal muscles of mice treated with sorafenib. Interventions that rescue or prevent these sorafenib-induced changes, such as taurine supplementation, may be helpful in attenuating sorafenib-induced cardiac injury.
- Published
- 2017
37. Atherosclerosis: Pathogenesis, Genetics and Experimental Models
- Author
-
Roberto Mota, Jonathon W. Homeister, Edward Moreira Bahnson, and Monte S. Willis
- Subjects
Pathology ,medicine.medical_specialty ,Heart disease ,business.industry ,Vascular disease ,Disease ,medicine.disease ,Bioinformatics ,Coronary artery disease ,Pathogenesis ,Atheroma ,medicine ,Endothelial dysfunction ,business ,Foam cell - Abstract
Atherosclerosis is a disease of the arteries that results in heart disease and stroke – the most common causes of death in developed countries, and a growing burden in developing countries. Atherosclerosis results from injury to the artery endothelium caused by mechanical and environmental factors, and the resulting inflammatory response in the vessel wall. The location and morphology of the atherosclerotic lesions predict the nature of the resulting vascular disease. Some risk factors for the disease are well known and current therapies are largely directed at modifying them, but the tendency to develop atherosclerosis is determined largely by polygenetic factors that are poorly understood. Recent developments in genetic studies have been applied to atherosclerosis, and are beginning to rapidly reveal the genetic factors that modulate the pathogenesis of this disease. Key Concepts: Atherosclerosis is a common, costly and deadly vascular disease that affects peoples of developed countries, and increasingly burdens developing countries. Atherosclerosis is an inflammatory disease of the arterial vascular wall. The pathogenesis of atherosclerosis is complex, but is generally explained by the ‘response to injury’ hypothesis. Atherosclerotic lesions have varied morphology, and clinical manifestations correlate with lesion type and location. Both environmental and heritable risk factors modulate atherosclerosis development. Candidate gene and linkage analysis studies have failed to identify previously unknown pathways in the pathogenesis of atherosclerosis. Recent publication of the HapMap has made possible genome-wide association studies aimed at probing the pathogenesis of atherosclerosis. Recent genome-wide association studies have reproducibly identified several loci involved in the pathogenesis of atherosclerosis, and most of the identified genes are newly implicated in the disease process. APOE- or LDLR-deficient mice are widely used models to study the pathogenesis of atherosclerosis. Keywords: atheroma; atherosclerosis; coronary artery disease; endothelial dysfunction; foam cell; hyperlipidaemia; hypertension; myocardial infarct; stroke; vascular inflammation
- Published
- 2017
38. BRG1 and BRM function antagonistically with c-MYC in adult cardiomyocytes to regulate conduction and contractility
- Author
-
Megan T. Quintana, Monte S. Willis, Joel S. Parker, Brian C. Jensen, Xin Chen, Mukesh K. Jain, Hyoung Gon Lee, Manasi Tannu, Darwin Jeyaraj, Scott J. Bultman, Zhongjing Wang, Julie A. Wolfram, and Darcy Holley
- Subjects
0301 basic medicine ,medicine.medical_specialty ,animal structures ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Biology ,Article ,law.invention ,Proto-Oncogene Proteins c-myc ,Contractility ,Electrocardiography ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Heart Conduction System ,law ,Internal medicine ,Cardiac conduction ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,Epigenetics ,Molecular Biology ,Heart Failure ,Oncogene ,Gene Expression Profiling ,DNA Helicases ,Nuclear Proteins ,medicine.disease ,Myocardial Contraction ,Phenotype ,SWI/SNF ,Cell biology ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,Heart failure ,Mutation ,Suppressor ,Cardiology and Cardiovascular Medicine ,Protein Binding ,Transcription Factors - Abstract
Rationale The contractile dysfunction that underlies heart failure involves perturbations in multiple biological processes ranging from metabolism to electrophysiology. Yet the epigenetic mechanisms that are altered in this disease state have not been elucidated. SWI/SNF chromatin-remodeling complexes are plausible candidates based on mouse knockout studies demonstrating a combined requirement for the BRG1 and BRM catalytic subunits in adult cardiomyocytes. Brg1 / Brm double mutants exhibit metabolic and mitochondrial defects and are not viable although their cause of death has not been ascertained. Objective To determine the cause of death of Brg1 / Brm double-mutant mice, to test the hypothesis that BRG1 and BRM are required for cardiac contractility, and to identify relevant downstream target genes. Methods and results A tamoxifen-inducible gene-targeting strategy utilizing αMHC-Cre-ERT was implemented to delete both SWI/SNF catalytic subunits in adult cardiomyocytes. Brg1 / Brm double-mutant mice were monitored by echocardiography and electrocardiography, and they underwent rapidly progressive ventricular dysfunction including conduction defects and arrhythmias that culminated in heart failure and death within 3 weeks. Mechanistically, BRG1/BRM repressed c - Myc expression, and enforced expression of a DOX-inducible c - MYC trangene in mouse cardiomyocytes phenocopied the ventricular conduction defects observed in Brg1 / Brm double mutants. BRG1/BRM and c-MYC had opposite effects on the expression of cardiac conduction genes, and the directionality was consistent with their respective loss- and gain-of-function phenotypes. To support the clinical relevance of this mechanism, BRG1/BRM occupancy was diminished at the same target genes in human heart failure cases compared to controls, and this correlated with increased c - MYC expression and decreased CX43 and SCN5A expression. Conclusion BRG1/BRM and c-MYC have an antagonistic relationship regulating the expression of cardiac conduction genes that maintain contractility, which is reminiscent of their antagonistic roles as a tumor suppressor and oncogene in cancer.
- Published
- 2017
39. 8th Annual rare disease scientific workshop: Evaluating early access models for patients: Flashpoints, frameworks & case studies for advancement: A summary
- Author
-
Bryan A. Wilson and Monte S. Willis
- Subjects
medicine.medical_specialty ,History ,Family medicine ,medicine ,Rare disease - Published
- 2017
40. The Head and the Heart
- Author
-
Monte S. Willis and Brian C. Jensen
- Subjects
0301 basic medicine ,03 medical and health sciences ,medicine.medical_specialty ,030104 developmental biology ,Head (linguistics) ,business.industry ,Internal medicine ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Connection (mathematics) - Published
- 2016
41. Bag3 P209L myopathies and efficacy of blocking signaling pathways with the therapeutic peptide, MMI-0100
- Author
-
Jessica M. Berthiaume, Monte S. Willis, and Ashley R. Hacker
- Subjects
Cardiac function curve ,business.industry ,Diastole ,Cardiomyopathy ,Ocean Engineering ,Protein degradation ,Pharmacology ,medicine.disease ,BAG3 ,Pathogenesis ,Fibrosis ,medicine ,Signal transduction ,business - Abstract
Background and Hypothesis: The Bcl2-associated anthanogene (BAG) 3 protein is a member of the BAG family of cochaperones, which play a critical role in cellular processes, including protein degradation and turnover. Over 30 Bag3 mutations have been identified, including a Proline209Leucine (P209L) missense mutation which causes a severe childhood cardiomyopathy. The mechanism by which Bag3 mutations causes cardiomyopathy is currently unknown, but the p38/MAPK signaling cascade has been shown to be altered in our animal model that coexpresses the human Bag3 P209L gene. We hypothesized the cell-permeant peptide, MMI-0100, which is known to inhibit MAPK-activated protein kinase 2 (MK2) activity in the p38/MAPK signaling cascade, would alleviate or reduce cardiac dysfunction. Experimental Design: Echocardiographic analysis of cardiac function of cardiac-specific Bag3 P209L transgenic (and wildtype littermate control) mice (20 – 22 months of age) was assessed by high-resolution ultrasound echocardiography (VisualSonics Vevo 2100, MS550D probe, cardiology package) to document the established disease-related cardiac dysfunction at baseline. Mice were then treated with 100mg/kg/day MMI-0100 nebulized daily for 30 days. Follow-up echocardiography was performed at 10, 20, and 30 days of MMI-0100 treatment. Echocardiographic analysis was performed to determine the systolic function (EF%, FS%), chamber dimensions, and wall thickness in systole and diastole using Vevo 2100 Workstation software package. Results: Blinded analysis of echocardiographic data identified that Bag3 P209L Tg+ mice had a baseline cardiac dysfunction compared to wildtype controls at 20 months of age (WT= 76% EF, 44% FS; Tg+= 66% EF, 36% FS). MMI-0100 treatment significantly attenuated this dysfunction by 20 days of MMI-0100 treatment (WT= 79% EF, 47% FS; Tg+= 80% EF, 48% FS), consistent with demonstrating for the first time MK2’s role in mediating p38 signaling in the pathogenesis of Bag3 P209L cardiomyopathy. Conclusion and Potential Impact: The MMI-0100 peptide has proven efficacious in several animal models of fibrosis driven by p38 signaling, as MK2 is a p38 downstream mediator. Future studies seek to translate the use of the MMI-0100 peptide in pediatric patients with Bag3 P209L cardiomyopathy through compassionate use FDA pathways.
- Published
- 2019
42. Identification of Metabolic Changes in Ileum, Jejunum, Skeletal Muscle, Liver, and Lung in a Continuous I.V. Pseudomonas aeruginosa Model of Sepsis Using Nontargeted Metabolomics Analysis
- Author
-
Nicolaas E. P. Deutz, Jessica M. Berthiaume, James R. Bain, Gabriella A. M. Ten Have, Amro Ilaiwy, Michael J. Muehlbauer, Monte S. Willis, Traci L. Parry, and Sara K. O’Neal
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Arginine ,Swine ,Ileum ,digestive system ,Article ,Pathology and Forensic Medicine ,Jejunum ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Metabolomics ,Pseudomonas Infections ,skin and connective tissue diseases ,Muscle, Skeletal ,Lung ,business.industry ,digestive, oral, and skin physiology ,Skeletal muscle ,Metabolism ,medicine.disease ,Small intestine ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Liver ,030220 oncology & carcinogenesis ,Pseudomonas aeruginosa ,Female ,sense organs ,business ,Metabolic Networks and Pathways - Abstract
Sepsis is a multiorgan disease affecting the ileum and jejunum (small intestine), liver, skeletal muscle, and lung clinically. The specific metabolic changes in the ileum, jejunum, liver, skeletal muscle, and lung have not previously been investigated. Live Pseudomonas aeruginosa, isolated from a patient, was given via i.v. catheter to pigs to induce severe sepsis. Eighteen hours later, ileum, jejunum, medial gastrocnemius skeletal muscle, liver, and lung were analyzed by nontargeted metabolomics analysis using gas chromatography/mass spectrometry. The ileum and the liver demonstrated significant changes in metabolites involved in linoleic acid metabolism: the ileum and lung had significant changes in the metabolism of valine/leucine/isoleucine; the jejunum, skeletal muscle, and liver had significant changes in arginine/proline metabolism; and the skeletal muscle and lung had significant changes in aminoacyl-tRNA biosynthesis, as analyzed by pathway analysis. Pathway analysis also identified changes in metabolic pathways unique for different tissues, including changes in the citric acid cycle (jejunum), β-alanine metabolism (skeletal muscle), and purine metabolism (liver). These findings demonstrate both overlapping metabolic pathways affected in different tissues and those that are unique to others and provide insight into the metabolic changes in sepsis leading to organ dysfunction. This may allow therapeutic interventions that focus on multiple tissues or single tissues once the relationship of the altered metabolites/metabolism to the underlying pathogenesis of sepsis is determined.
- Published
- 2019
43. Walk the Line: The Role of Ubiquitin in Regulating Transcription in Myocytes
- Author
-
Monte S. Willis and Vidyani Suryadevara
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Transcription, Genetic ,Physiology ,Ubiquitin-Protein Ligases ,Review ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Transcription (biology) ,medicine ,Myocyte ,Animals ,Humans ,Muscle, Skeletal ,Transcription factor ,Muscle Cells ,biology ,Skeletal muscle ,Ubiquitin ligase ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
The ubiquitin-proteasome offers novel targets for potential therapies with their specific activities and tissue localization. Recently, the expansion of our understanding of how ubiquitin ligases (E3s) specifically regulate transcription has demonstrated their roles in skeletal muscle, complementing their roles in protein quality control and protein degradation. This review focuses on skeletal muscle E3s that regulate transcription factors critical to myogenesis and the maintenance of skeletal muscle wasting diseases.
- Published
- 2019
44. Cardiomyocyte glucocorticoid and mineralocorticoid receptors directly and antagonistically regulate heart disease in mice
- Author
-
Pierre Chambon, Julie F. Foley, Robert H. Oakley, John A. Cidlowski, Bo He, Xiaojiang Xu, Monte S. Willis, Diana Cruz-Topete, Celso E. Gomez-Sanchez, Page Myers, National Institute of Environmental Health Sciences [Durham] (NIEHS-NIH), National Institutes of Health [Bethesda] (NIH), University of Mississippi Medical Center (UMMC), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of North Carolina [Chapel Hill] (UNC), University of North Carolina System (UNC), and univOAK, Archive ouverte
- Subjects
medicine.medical_specialty ,Heart Diseases ,Heart disease ,medicine.drug_class ,Cardiomegaly ,Mice, Transgenic ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Biochemistry ,Article ,03 medical and health sciences ,Mice ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Glucocorticoid receptor ,Mineralocorticoid receptor ,Receptors, Glucocorticoid ,Internal medicine ,medicine ,Humans ,Animals ,Myocytes, Cardiac ,Calcium Signaling ,Glucocorticoids ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Ventricular Remodeling ,business.industry ,Cell Biology ,medicine.disease ,Endocrinology ,Receptors, Mineralocorticoid ,Nuclear receptor ,Mineralocorticoid ,Heart failure ,Calcium ,business ,Oxidative stress ,Glucocorticoid ,Gene Deletion ,Signal Transduction ,medicine.drug - Abstract
Stress is increasingly associated with heart dysfunction and is linked to higher mortality rates in patients with cardiometabolic disease. Glucocorticoids are primary stress hormones that regulate homeostasis through two nuclear receptors, the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR), both of which are present in cardiomyocytes. To examine the specific and coordinated roles that these receptors play in mediating the direct effects of stress on the heart, we generated mice with cardiomyocyte-specific deletion of GR (cardioGRKO), MR (cardioMRKO), or both GR and MR (cardioGRMRdKO). The cardioGRKO mice spontaneously developed cardiac hypertrophy and left ventricular systolic dysfunction and died prematurely from heart failure. In contrast, the cardioMRKO mice exhibited normal heart morphology and function. Surprisingly, despite the presence of myocardial stress, the cardioGRMRdKO mice were resistant to the cardiac remodeling, left ventricular dysfunction, and early death observed in the cardioGRKO mice. Gene expression analysis revealed the loss of gene changes associated with impaired Ca(2+) handling, increased oxidative stress, and enhanced cell death and the presence of gene changes that limited the hypertrophic response and promoted cardiomyocyte survival in the double knockout hearts. Re-expression of MR in cardioGRMRdKO hearts reversed many of the cardioprotective gene changes and resulted in cardiac failure. These findings reveal a critical role for balanced cardiomyocyte GR and MR stress signaling in cardiovascular health. Therapies that shift stress signaling in the heart to favor more GR and less MR activity may provide an improved approach for treating heart disease.
- Published
- 2019
45. Doxorubicin Exposure Causes Subacute Cardiac Atrophy Dependent on the Striated Muscle-Specific Ubiquitin Ligase MuRF1
- Author
-
Wei Huang, Ralph B. D'Agostino, W. Gregory Hundley, Cynthia Zhou, Traci L. Parry, Roberto Mota, Sean T Hicks, David I. Brown, Monte S. Willis, Brian C. Jensen, Ju Youn Beak, Jennifer H. Jordan, Melissa C. Caughey, and Michael Sola
- Subjects
Male ,Gene Expression ,Muscle Proteins ,030204 cardiovascular system & hematology ,Ligases ,Tripartite Motif Proteins ,Mice ,0302 clinical medicine ,Anthracyclines ,Myocytes, Cardiac ,0303 health sciences ,Dilated cardiomyopathy ,Heart ,Magnetic Resonance Imaging ,Muscle atrophy ,Up-Regulation ,Muscular Atrophy ,Echocardiography ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Injections, Intraperitoneal ,medicine.drug ,Cardiac function curve ,medicine.medical_specialty ,Anthracycline ,Ubiquitin-Protein Ligases ,Antineoplastic Agents ,Article ,03 medical and health sciences ,Atrophy ,Internal medicine ,medicine ,Animals ,Humans ,Doxorubicin ,030304 developmental biology ,Heart Failure ,Cardiotoxicity ,Dose-Response Relationship, Drug ,business.industry ,Ubiquitin ,Myocardium ,medicine.disease ,Muscle, Striated ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,Heart failure ,business - Abstract
Background Anthracycline chemotherapeutics, such as doxorubicin, are used widely in the treatment of numerous malignancies. The primary dose-limiting adverse effect of anthracyclines is cardiotoxicity that often presents as heart failure due to dilated cardiomyopathy years after anthracycline exposure. Recent data from animal studies indicate that anthracyclines cause cardiac atrophy. The timing of onset and underlying mechanisms are not well defined, and the relevance of these findings to human disease is unclear. Methods and Results Wild-type mice were sacrificed 1 week after intraperitoneal administration of doxorubicin (1–25 mg/kg), revealing a dose-dependent decrease in cardiac mass ( R 2 =0.64; P 2 ; P 2 =0.91; P =0.003) and a molecular profile of muscle atrophy. To investigate the determinants of doxorubicin-induced cardiac atrophy, we administered doxorubicin 20 mg/kg to mice lacking MuRF1 (MuRF1 −/− ) and wild-type littermates. MuRF1 −/− mice were protected from cardiac atrophy and exhibited no reduction in contractile function. To explore the clinical relevance of these findings, we analyzed cardiac magnetic resonance imaging data from 70 patients in the DETECT-1 cohort and found that anthracycline exposure was associated with decreased cardiac mass evident within 1 month and persisting to 6 months after initiation. Conclusions Doxorubicin causes a subacute decrease in cardiac mass in both mice and humans. In mice, doxorubicin-induced cardiac atrophy is dependent on MuRF1. These findings suggest that therapies directed at preventing or reversing cardiac atrophy might preserve the cardiac function of cancer patients receiving anthracyclines.
- Published
- 2019
46. The sympathetic nervous system regulates skeletal muscle motor innervation and acetylcholine receptor stability
- Author
-
Tan Zhang, D. Clark Files, Osvaldo Delbono, Ronald W. Oppenheim, Meaghan O’Meara, Akiva Mintz, Ping Kwan, Martín Carlos Abba, Elsa Idaliz Silva Lopez, Cristina M. Furdui, Anna Carolina Zaia Rodrigues, Alexander Birbrair, Zhong-Min Wang, Monte S. Willis, Andrea S. Pereyra, and María Laura Messi
- Subjects
0301 basic medicine ,Sympathetic nervous system ,animal structures ,Physiology ,SYMPATHETIC NERVOUS SYSTEM ,Motor nerve ,Skeletal muscle ,Neuromuscular junction ,030204 cardiovascular system & hematology ,Synaptic Transmission ,Article ,Ciencias Biológicas ,purl.org/becyt/ford/1 [https] ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Postsynaptic potential ,NEUROMUSCULAR JUNCTION ,medicine ,Animals ,Receptors, Cholinergic ,MUSCLE DENERVATION ,Muscle, Skeletal ,purl.org/becyt/ford/1.6 [https] ,Motor Neurons ,Muscle denervation ,Muscle Denervation ,business.industry ,Muscle weakness ,Bioquímica y Biología Molecular ,Muscle atrophy ,Acetylcholine ,Muscle innervation ,Muscular Atrophy ,030104 developmental biology ,medicine.anatomical_structure ,Ciencias Médicas ,medicine.symptom ,MUSCLE INNERVATION ,business ,SKELETAL MUSCLE ,Neuroscience ,CIENCIAS NATURALES Y EXACTAS - Abstract
Aim: Symptoms of autonomic failure are frequently the presentation of advanced age and neurodegenerative diseases that impair adaptation to common physiologic stressors. The aim of this work was to examine the interaction between the sympathetic and motor nervous system, the involvement of the sympathetic nervous system (SNS) in neuromuscular junction (NMJ) presynaptic motor function, the stability of postsynaptic molecular organization, and the skeletal muscle composition and function. Methods: Since muscle weakness is a symptom of diseases characterized by autonomic dysfunction, we studied the impact of regional sympathetic ablation on muscle motor innervation by using transcriptome analysis, retrograde tracing of the sympathetic outflow to the skeletal muscle, confocal and electron microscopy, NMJ transmission by electrophysiological methods, protein analysis, and state of the art microsurgical techniques, in C57BL6, MuRF1KO and Thy-1 mice. Results: We found that the SNS regulates motor nerve synaptic vesicle release, skeletal muscle transcriptome, muscle force generated by motor nerve activity, axonal neurofilament phosphorylation, myelin thickness, and myofibre subtype composition and CSA. The SNS also modulates the levels of postsynaptic membrane acetylcholine receptor by regulating the Gαi2 -Hdac4-Myogenin-MuRF1pathway, which is prevented by the overexpression of the guanine nucleotide-binding protein Gαi2 (Q205L), a constitutively active mutant G protein subunit. Conclusion: The SNS regulates NMJ transmission, maintains optimal Gαi2 expression, and prevents any increase in Hdac4, myogenin, MuRF1, and miR-206. SNS ablation leads to upregulation of MuRF1, muscle atrophy, and downregulation of postsynaptic AChR. Our findings are relevant to clinical conditions characterized by progressive decline of sympathetic innervation, such as neurodegenerative diseases and aging., Centro de Investigaciones Inmunológicas Básicas y Aplicadas
- Published
- 2019
47. Fibrosis in Disease : An Organ-Based Guide to Disease Pathophysiology and Therapeutic Considerations
- Author
-
Monte S. Willis, Cecelia C. Yates, Jonathan C. Schisler, Monte S. Willis, Cecelia C. Yates, and Jonathan C. Schisler
- Subjects
- Fibrosis
- Abstract
Fibroproliferative diseases are a broad spectrum of entities from organ-specific involvement (e.g., pulmonary, heart, liver, and kidney fibrosis) to multi-system diseases such as systemic sclerosis and sclerodermatous graft vs. host disease. These diseases also encompass pathophysiologies not readily recognizably related, such as macular degeneration and cancer metastasis. Fibroproliferative diseases are a leading cause of morbidity and mortality and can affect all tissues and organ systems. Remarkable progress in elucidating the pathogenesis of these common diseases with fibrotic components, including the critical roles of myofibroblasts and the molecular mechanisms driving the transcriptional activation involved in the induction of fibrosis. As the importance of these processes is realized in the long-term recovery and treatment of diseases, effective anti-fibrotic therapies targeting the underlying ongoing disease processes are lacking. The complexity of discovering and applying therapies to fibroproliferative disease may be due to the diversity of the systems the pathogenesis of disease itself involves. By nature, fibroproliferative diseases are interdisciplinary, involving multiple cell types (organ-specific epithelial cells), immune cells, endothelial cells, and fibroblasts. Bone marrow, cytokines, and organ-specific pathologies further speckle both the clinical and scientific disciplines in such a way that communication is often limited to the clinical or scientific tribes we live in, despite the greatest access to information known to man available today. Therefore, the primary focus of this text is to bring together authors from a diversity of both clinical, scientific, and therapeutic backgrounds for readers to more fully appreciate that fantastic platform that is available to build upon to lessen the isolation of the clinical and scientific disciplines. With advances in the discovery of pre-clinical therapeutic targets (at least 20+ to date) involving TGF-beta (and other cytokines), transcription factors, and downstream kinases, it's important to both recognize the broader impact and potential opportunities that exist even today. This book will serve as a state-of-the-art resource for physicians and translational medical researchers alike who are interested in the rapidly evolving field of fibroproliferative diseases. The book will provide new insight into the fundamental mechanisms of classic fibrotic pathophysiologic processes like myocardial infarction, idiopathic pulmonary fibrosis, chronic kidney disease, wound healing, and systemic sclerosis. It will also highlight the many new areas of therapeutic investigation currently underway. Lastly, we will touch upon newly emerging fields investigating the role of fibrosis in macular degeneration and cancer metastasis. The chapters will be written by established experts in their fields, including clinicians (cardiologists, cardiovascular surgeons, pathologists, and general practitioners) and translational biomedical researchers in a wide range of disciplines. However, the material will certainly have a broader audience including medical residents, fellows, and general practitioners as well as M.D. or Ph.D. post-doctoral research fellows. While comprehensive, we'll attempt to present the material in a manner that simplifies the complex pathophysiologic mechanisms that underlie common fibroproliferative diseases while making it appealing to a broad audience.
- Published
- 2019
48. Nebulized Delivery of the MAPKAP Kinase 2 Peptide Inhibitor MMI-0100 Protects Against Ischemia-Induced Systolic Dysfunction
- Author
-
Megan T. Quintana, Cynthia Lander, Brian C. Cooley, David I. Brown, and Monte S. Willis
- Subjects
0301 basic medicine ,Cardiac function curve ,Myocarditis ,p38 mitogen-activated protein kinases ,Ischemia ,Bioengineering ,Inflammation ,030204 cardiovascular system & hematology ,Pharmacology ,Biochemistry ,Article ,Analytical Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,Drug Discovery ,medicine ,Myocardial infarction ,business.industry ,medicine.disease ,030104 developmental biology ,Anesthesia ,Molecular Medicine ,medicine.symptom ,Ligation ,business - Abstract
Acute myocardial infarction (AMI) results in systolic dysfunction, myocarditis and fibrotic remodeling, which causes irreversible pathological remodeling of the heart. Associated cell death and inflammation cause cytokine release, which activates the p38 MAPK signaling pathway to propagate damaging signals via MAPKAP kinase 2 (MK2). Previously we showed that intraperitoneal injection of a cell permeable peptide inhibitor of MK2, MMI-0100, protects against fibrosis, apoptosis and systolic dysfunction in a mouse model of AMI induced by left-anterior descending coronary artery (LAD) ligation. Here we tested a new route of administration of MMI-0100: inhalation of nebulized peptide. When given within 30 min of AMI and daily for 2 weeks thereafter, both inhaled and injected MMI-0100 improved cardiac function as measured by conscious echocardiography. Limited fibrosis was observed after 2 weeks by Massons trichrome staining, suggesting that MMI-0100 protects the heart prior to the formation of significant fibrosis. These results support a nebulized route of administration of MMI-0100 can protect the myocardium from ischemic damage.
- Published
- 2016
49. Alzheimer’s Disease, heart failure, and musculoskeletal defects and their relationship to clinical co‐morbidities
- Author
-
Monte S. Willis
- Subjects
medicine.medical_specialty ,business.industry ,Disease ,medicine.disease ,Biochemistry ,Heart failure ,Internal medicine ,Genetics ,medicine ,Cardiology ,Co morbidity ,business ,Molecular Biology ,Biotechnology - Published
- 2020
50. Functional analysis of cardiac‐specific loss of MuRF1 in diabetic cardiomyopathy
- Author
-
Michael Klüppel, Sahith Rajalingam, and Monte S. Willis
- Subjects
medicine.medical_specialty ,business.industry ,Diabetic cardiomyopathy ,Internal medicine ,Genetics ,medicine ,Cardiology ,medicine.disease ,business ,Molecular Biology ,Biochemistry ,Functional analysis (psychology) ,Biotechnology - Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.