cardiac sarcomere
Recently Published Documents


TOTAL DOCUMENTS

41
(FIVE YEARS 6)

H-INDEX

14
(FIVE YEARS 1)

2021 ◽  
Vol 22 (21) ◽  
pp. 11368
Author(s):  
Xixia Peng ◽  
Gang Feng ◽  
Yanyong Zhang ◽  
Yuhua Sun

Cardiac development is a complex process that is strictly controlled by various factors, including PcG protein complexes. Several studies have reported the critical role of PRC2 in cardiogenesis. However, little is known about the regulation mechanism of PRC1 in embryonic heart development. To gain more insight into the mechanistic role of PRC1 in cardiogenesis, we generated a PRC1 loss-of-function zebrafish line by using the CRISPR/Cas9 system targeting rnf2, a gene encoding the core subunit shared by all PRC1 subfamilies. Our results revealed that Rnf2 is not involved in cardiomyocyte differentiation and heart tube formation, but that it is crucial to maintaining regular cardiac contraction. Further analysis suggested that Rnf2 loss-of-function disrupted cardiac sarcomere assembly through the ectopic activation of non-cardiac sarcomere genes in the developing heart. Meanwhile, Rnf2 deficiency disrupts the construction of the atrioventricular canal and the sinoatrial node by modulating the expression of bmp4 and other atrioventricular canal marker genes, leading to an impaired cardiac conduction system. The disorganized cardiac sarcomere and defective cardiac conduction system together contribute to defective cardiac contraction. Our results emphasize the critical role of PRC1 in the cardiac development.


Author(s):  
Claudia Crocini ◽  
Michael Gotthardt

AbstractThe sarcomere is the fundamental structural and functional unit of striated muscle and is directly responsible for most of its mechanical properties. The sarcomere generates active or contractile forces and determines the passive or elastic properties of striated muscle. In the heart, mutations in sarcomeric proteins are responsible for the majority of genetically inherited cardiomyopathies. Here, we review the major determinants of cardiac sarcomere mechanics including the key structural components that contribute to active and passive tension. We dissect the molecular and structural basis of active force generation, including sarcomere composition, structure, activation, and relaxation. We then explore the giant sarcomere-resident protein titin, the major contributor to cardiac passive tension. We discuss sarcomere dynamics exemplified by the regulation of titin-based stiffness and the titin life cycle. Finally, we provide an overview of therapeutic strategies that target the sarcomere to improve cardiac contraction and filling.


Cell Reports ◽  
2020 ◽  
Vol 32 (9) ◽  
pp. 108090
Author(s):  
Sharissa L. Latham ◽  
Nadine Weiß ◽  
Kristin Schwanke ◽  
Claudia Thiel ◽  
David R. Croucher ◽  
...  

2020 ◽  
Vol 15 (4) ◽  
pp. 457-469 ◽  
Author(s):  
Joshua B. Holmes ◽  
Chang Yoon Doh ◽  
Ranganath Mamidi ◽  
Jiayang Li ◽  
Julian E. Stelzer

2019 ◽  
Vol 125 (Suppl_1) ◽  
Author(s):  
Anthony M PETTINATO ◽  
Feria Ladha ◽  
Ketan Thakar ◽  
Travis Hinson
Keyword(s):  

2018 ◽  
Vol 115 (26) ◽  
pp. 6727-6732 ◽  
Author(s):  
Caralynn M. Wilczewski ◽  
Austin J. Hepperla ◽  
Takashi Shimbo ◽  
Lauren Wasson ◽  
Zachary L. Robbe ◽  
...  

Cardiac development relies on proper cardiomyocyte differentiation, including expression and assembly of cell-type-specific actomyosin subunits into a functional cardiac sarcomere. Control of this process involves not only promoting expression of cardiac sarcomere subunits but also repressing expression of noncardiac myofibril paralogs. This level of transcriptional control requires broadly expressed multiprotein machines that modify and remodel the chromatin landscape to restrict transcription machinery access. Prominent among these is the nucleosome remodeling and deacetylase (NuRD) complex, which includes the catalytic core subunit CHD4. Here, we demonstrate that direct CHD4-mediated repression of skeletal and smooth muscle myofibril isoforms is required for normal cardiac sarcomere formation, function, and embryonic survival early in gestation. Through transcriptomic and genome-wide analyses of CHD4 localization, we identified unique CHD4 binding sites in smooth muscle myosin heavy chain, fast skeletal α-actin, and the fast skeletal troponin complex genes. We further demonstrate that in the absence of CHD4, cardiomyocytes in the developing heart form a hybrid muscle cell that contains cardiac, skeletal, and smooth muscle myofibril components. These misexpressed paralogs intercalate into the nascent cardiac sarcomere to disrupt sarcomere formation and cause impaired cardiac function in utero. These results demonstrate the genomic and physiological requirements for CHD4 in mammalian cardiac development.


2018 ◽  
Vol 44 (1) ◽  
pp. 87-96.e5 ◽  
Author(s):  
Anant Chopra ◽  
Matthew L. Kutys ◽  
Kehan Zhang ◽  
William J. Polacheck ◽  
Calvin C. Sheng ◽  
...  

2016 ◽  
Vol 12 (10) ◽  
pp. e1005126 ◽  
Author(s):  
Lauren J. Dupuis ◽  
Joost Lumens ◽  
Theo Arts ◽  
Tammo Delhaas

Sign in / Sign up

Export Citation Format

Share Document