You are on page 1of 23

Myosin binding protein C, cardiac

The myosin-binding protein C,


MYBPC3
cardiac-type is a protein that in
humans is encoded by the
MYBPC3 gene.[5] This isoform is
expressed exclusively in heart
muscle during human and mouse
development,[6] and is distinct
from those expressed in slow
skeletal muscle (MYBPC1) and
fast skeletal muscle (MYBPC2).

Available structures
PDB Ortholog search: PDBe (https://www.ebi.ac.uk/pdbe/search
Contents Results.html?display=both&term=O70468%20or%20Q1489
Structure 6%20or%20A5YM48) RCSB (http://www.rcsb.org/pdb/searc
Function h/smartSubquery.do?smartSearchSubtype=UpAccessionId
Query&accessionIdList=O70468,Q14896,A5YM48)
Genetics
List of PDB id codes
Pathomechanisms
Therapy 1GXE (https://www.rcsb.org/structure/1GXE), 2AVG
(https://www.rcsb.org/structure/2AVG), 2K1M (http
Notes
s://www.rcsb.org/structure/2K1M), 2MQ0 (https://w
References ww.rcsb.org/structure/2MQ0), 2MQ3 (https://www.rc
Further reading sb.org/structure/2MQ3), 2V6H (https://www.rcsb.or
External links g/structure/2V6H), 3CX2 (https://www.rcsb.org/stru
cture/3CX2), 1PD6 (https://www.rcsb.org/structure/
1PD6)
Structure
Identifiers
cMyBP-C is a 140.5 kDa protein
composed of 1273 amino Aliases MYBPC3 (https://www.genenames.org/cgi-bin/gene_symb
acids.[7][8][9] cMyBP-C is a ol_report?hgnc_id=7551), CMD1MM, CMH4, FHC,
myosin-associated protein that LVNC10, MYBP-C, Myosin binding protein C, cardiac,
binds at 43 nm intervals along the cMyBP-C, myosin binding protein C3
myosin thick filament backbone, External OMIM: 600958 (https://omim.org/entry/600958) MGI:
stretching for 200 nm on either IDs 102844 (http://www.informatics.jax.org/marker/MGI:10284
side of the M-line within the 4) HomoloGene: 215 (https://www.ncbi.nlm.nih.gov/entre
crossbridge-bearing zone (C- z/query.fcgi?cmd=Retrieve&db=homologene&dopt=Homol
region) of the A band in striated oGene&list_uids=215) GeneCards: MYBPC3 (https://ww
muscle.[10] The approximate w.genecards.org/cgi-bin/carddisp.pl?gene=MYBPC3)
stoichiometry of cMyBP-C along Gene location (Human)
the thick filament is 1 per 9-10
myosin molecules, or 37 cMyBP-
C molecules per thick
filament.[11] In addition to
myosin, cMyBP-C also binds titin
and actin.[12][13] The cMyBP-C
isoform expressed in cardiac
muscle differs from those
expressed in slow and fast skeletal Chr. Chromosome 11 (human)[1]
muscle (MYBPC1 and MYBPC2,
respectively) by three features: (1)
an additional immunoglobulin
(Ig)-like domain on the N- Band 11p11.2 Start 47,331,406 bp[1]
terminus, (2) a linker region End 47,352,702 bp[1]
between the second and third Ig
domains, and (3) an additional Gene location (Mouse)
loop in the sixth Ig domain.[14]
cMyBP-C appears necessary for
normal order, filament length and
lattice spacing within the structure
of the sarcomere.[15][16]
Chr. Chromosome 2 (mouse)[2]
Function
cMyBP-C is not essential for
sarcomere formation during
embryogenesis, but is crucial for Band 2|2 E1 Start 91,118,144 bp[2]
sarcomere organization and End 91,136,516 bp[2]
maintenance of normal cardiac RNA expression pattern
function. Absence of cMyBP-C
(Mybpc3-targeted knock-out
mice) results in severe cardiac
hypertrophy, increased heart-
weight-to-body-weight-ratios,
enlargement of ventricles,
increased myofilament Ca2+
sensitivity and depressed diastolic
and systolic function.[17][18][19]
Histologically, Mybpc3-targeted More reference expression data (http://biogps.org/gene/4607/)
knock-out hearts display
structural rearrangements with Gene ontology
cardiac myocyte disarray and Molecular • myosin binding (http://amigo.geneontology.org/amigo/
increased interstitial fibrosis function term/GO:0017022)
similar to patients with • titin binding (http://amigo.geneontology.org/amigo/ter
hypertrophic cardiomyopathy, m/GO:0031432)
without obvious alterations in • metal ion binding (http://amigo.geneontology.org/ami
shape or size of single cardiac go/term/GO:0046872)
myocytes. Ultrastructural • myosin heavy chain binding (http://amigo.geneontolo
examination revealed a loss of gy.org/amigo/term/GO:0032036)
lateral alignment of adjacent • GO:0001948 protein binding (http://amigo.geneontolo
myofibrils with their Z-lines gy.org/amigo/term/GO:0005515,)
misaligned.[17][18][20][21] • identical protein binding (http://amigo.geneontology.or
g/amigo/term/GO:0042802)
cMyBP-C appears to act as a • actin binding (http://amigo.geneontology.org/amigo/te
brake on cardiac contraction, as rm/GO:0003779)
loaded shortening, power and
• ATPase activator activity (http://amigo.geneontology.
cycling kinetics all increase in
org/amigo/term/GO:0001671)
cMyBP-C knockout mice.[22]
• structural constituent of muscle (http://amigo.geneont
Consistent with this notion,
ology.org/amigo/term/GO:0008307)
cMyBP-C knockout mice exhibit
• actin filament binding (http://amigo.geneontology.org/
an abnormal systolic timecourse,
amigo/term/GO:0051015)
with a shortened elastance
• muscle alpha-actinin binding (http://amigo.geneontolo
timecourse and lower peak
gy.org/amigo/term/GO:0051371)
elastance in vivo,[23] and an
• structural molecule activity conferring elasticity (http://
accelerated force development in
amigo.geneontology.org/amigo/term/GO:0097493)
isolated, skinned cardiac fibers[24]
suggesting that cMyBP-C is Cellular • cytosol (http://amigo.geneontology.org/amigo/term/G
required to constrain the component O:0005829)
crossbridges in order to sustain a • C zone (http://amigo.geneontology.org/amigo/term/G
normal ejection. O:0014705)
• sarcomere (http://amigo.geneontology.org/amigo/ter
cMyBP-C regulates the m/GO:0030017)
positioning of myosin and actin • striated muscle myosin thick filament (http://amigo.ge
for interaction and acts as a tether neontology.org/amigo/term/GO:0005863)
to the myosin S1 heads, limiting • myosin filament (http://amigo.geneontology.org/amig
their mobility. This results in a o/term/GO:0032982)
decreased number of crossbridges • A band (http://amigo.geneontology.org/amigo/term/G
formed, which hinders force O:0031672)
generation, due to its N-terminal • Z disc (http://amigo.geneontology.org/amigo/term/G
C1-M-C2 region interacting with O:0030018)
the myosin-S2 • M band (http://amigo.geneontology.org/amigo/term/G
domain. [25][26][27][28]
O:0031430)
Furthermore, cMyBP-C
• cardiac myofibril (http://amigo.geneontology.org/amig
contributes to the regulation of
o/term/GO:0097512)
cardiac contraction at short
• striated muscle thin filament (http://amigo.geneontolo
sarcomere length and is required
gy.org/amigo/term/GO:0005865)
for complete relaxation in
diastole.[19][29] Biological • regulation of muscle filament sliding (http://amigo.gen
process eontology.org/amigo/term/GO:0032971)
Interactions of cMyBP-C with its • positive regulation of ATPase activity (http://amigo.ge
binding partners vary with its neontology.org/amigo/term/GO:0032781)
posttranslational modification • cardiac muscle contraction (http://amigo.geneontolog
status. At least three extensively y.org/amigo/term/GO:0060048)
characterized phosphorylation • heart morphogenesis (http://amigo.geneontology.org/
sites (Ser273, 282 and 302; amigo/term/GO:0003007)
numbering refers to the mouse • ventricular cardiac muscle tissue morphogenesis (htt
sequence) are localized in the M p://amigo.geneontology.org/amigo/term/GO:0055010)
motif of cMyBP-C and are • regulation of striated muscle contraction (http://amig
targeted by protein kinases in a o.geneontology.org/amigo/term/GO:0006942)
hierarchical order of events. In its • cell adhesion (http://amigo.geneontology.org/amigo/te
dephosphorylated state, cMyBP-C rm/GO:0007155)
binds predominantly to myosin S2 • muscle filament sliding (http://amigo.geneontology.or
and brakes crossbridge formation, g/amigo/term/GO:0030049)
however, when phosphorylated in • striated muscle contraction (http://amigo.geneontolog
response to β-adrenergic y.org/amigo/term/GO:0006941)
stimulation through activating • actin filament organization (http://amigo.geneontolog
cAMP-dependent protein kinase y.org/amigo/term/GO:0007015)
(PKA), it favours binding to actin, • sarcomere organization (http://amigo.geneontology.or
then accelerating crossbridge g/amigo/term/GO:0045214)
formation, enhancing force • striated muscle myosin thick filament assembly (htt
development and promoting p://amigo.geneontology.org/amigo/term/GO:0071688)
relaxation. [30] Protein kinases • muscle contraction (http://amigo.geneontology.org/a
identified thus far to migo/term/GO:0006936)
phosphorylate cMyBP-C in the M
• skeletal muscle thin filament assembly (http://amigo.g
motif are PKA,[31][32][33][34][35] eneontology.org/amigo/term/GO:0030240)
Ca2+/calmodulin-dependent
• skeletal muscle myosin thick filament assembly (htt
kinase II (CaMKII),[36] ribosomal
p://amigo.geneontology.org/amigo/term/GO:0030241)
s6 kinase (RSK),[37]protein kinase
• cardiac muscle fiber development (http://amigo.gene
D (PKD),[38][39] and protein
ontology.org/amigo/term/GO:0048739)
kinase C (PKC).[34] Furthermore,
• cardiac myofibril assembly (http://amigo.geneontolog
GSK3β was described as another
y.org/amigo/term/GO:0055003)
protein kinase to phosphorylate
• cardiac muscle tissue morphogenesis (http://amigo.g
cMyBP-C outside the M-domain
eneontology.org/amigo/term/GO:0055008)
in the proline-alanine-rich actin-
binding site at Ser133 in human Sources:Amigo (http://amigo.geneontology.org/) / QuickGO (https://
myocardium (mouse Ser131).[40] www.ebi.ac.uk/QuickGO/)
Phosphorylation is required for Orthologs
normal cardiac function and
Species Human Mouse
cMyBP-C stability,[41][42] and
Entrez
overall phosphorylation levels of 4607 (https://www.ncb 17868 (https://www.ncbi.
cMyBP-C are reduced in human i.nlm.nih.gov/entrez/q nlm.nih.gov/entrez/query.
and experimental heart uery.fcgi?db=gene&c fcgi?db=gene&cmd=retri
failure. [43][44] Other md=retrieve&dopt=def eve&dopt=default&list_ui
posttranslational modifications of ault&list_uids=4607&r ds=17868&rn=1)
cMyBP-C exist, which occur n=1)
throughout the protein and are not
thoroughly characterised yet, such Ensembl
as acetylation,[45] ENSG00000134571 ENSMUSG00000002100
citrullination,[46] S- (http://www.ensembl.o (http://www.ensembl.org/
glutathiolation, [47][48][49][50] S- rg/Homo_sapiens/gen Mus_musculus/genevie
nitrosylation [51] and eview?gene=ENSG00 w?gene=ENSMUSG000
carbonylation. [52] 000134571;db=core) 00002100;db=core)

Genetics UniProt
The cloning of the human Q14896 (https://www. O70468 (https://www.uni
MYBPC3 cDNA and localization uniprot.org/uniprot/Q1 prot.org/uniprot/O70468)
of the gene on human 4896)
chromosome 11p11.2 has assisted
the structure and function of RefSeq
cMyBP-C.[53] MYBPC3 became (mRNA)
NM_000256 (https://w NM_008653 (https://ww
therefore the “best” candidate ww.ncbi.nlm.nih.gov/e w.ncbi.nlm.nih.gov/entre
gene for the CMH4 locus for ntrez/viewer.fcgi?val= z/viewer.fcgi?val=NM_00
hypertrophic cardiomyopathy that NM_000256) 8653)
was initially mapped by the group
of Schwartz.[54] MYBPC3 RefSeq
NP_000247 (https://w n/a
mutations segregating in families (protein)
ww.ncbi.nlm.nih.gov/e
with hypertrophic
ntrez/viewer.fcgi?val=
cardiomyopathy have been
[55][56] NP_000247)
identified. MYBPC3 was
NP_000247.2 (https://
thus the fourth gene for
www.ncbi.nlm.nih.gov/
hypertrophic cardiomyopathy,
entrez/viewer.fcgi?val
following MYH7, encoding β-
=NP_000247.2)
myosin heavy chain, TNNT2 and
TPM1, encoding cardiac troponin
Location Chr 11: 47.33 – 47.35 Mb Chr 2: 91.12 – 91.14 Mb (http
T and α-tropomyosin,
(UCSC) (https://genome.ucsc.edu/ s://genome.ucsc.edu/cgi-bin/
respectively, earmarking
cgi-bin/hgTracks?org=Hu hgTracks?org=Mouse&db=m
hypertrophic cardiomyopathy as a
man&db=hg38&position=c m0&position=chr2:91118144-
disease of the sarcomere.
hr11:47331406-47352702) 91136516)
To date, roughly 350 mutations in PubMed [3] [4]

MYBPC3 have been identified, search


and in large part, the mutations Wikidata
result in protein truncation, shifts
View/Edit Human View/Edit Mouse
in reading frames, and premature
termination codons.[57][58]
Genetic studies have revealed significant overlap between genotypes and phenotypes as MYBPC3
mutations can lead to various forms of cardiomyopathies, such as dilated cardiomyopathy[59] and left
ventricular noncompaction cardiomyopathy.[60] In patients with isolated or familial cases of dilated
cardiomyoathy, MYBPC3 mutations represented the second highest number of known mutations.[59]
Furthermore, a 25-bp intronic MYBPC3 deletion leading to protein truncation is present in 4% of the
population in South India and is associated with a higher risk to develop heart failure.[61] Founder
MYBPC3 mutations have been reported in Iceland, Italy, The Netherlands, Japan, France and Finland,
where they represent a large percentage of cases with hypertrophic cardiomyopathy. All of them are
truncating mutations, resulting in a shorter protein, lacking the regulatory phosphorylatable M motif
and/or major binding domains to other sarcomeric proteins.[62][63][64][65][66][67][68] A body of evidence
indicates that patients with more than 1 mutation often develop a more severe phenotype,[69] and a
significant fraction of childhood-onset hypertrophic cardiomyopathy(14%) is caused by compound
genetic variants.[70] This suggests that a gene-dosage effect might be responsible for manifestations at a
younger age. A total of 51 cases of homozygotes or compound heterozygotes have been reported, most of
them with double truncating MYBPC3 mutations and associated with severe cardiomyopathy, leading to
heart failure and death within the first year of life.[71]
Pathomechanisms
A great understanding of how MYBPC3 mutations lead to the development of inherited cardiomyopathy
came from the analyses of human myocardial samples, gene transfer in different cell lines, naturally-
occurring or transgenic animal models and more recently disease modeling using induced pluripotent
stem cells (iPSC)-derived cardiac myocytes.[72][73] Although access to human myocardial samples is
difficult, at least some studies provided evidence that truncated cMyBP-Cs, resulting from truncating
MYBPC3 mutations are not detectable in human patient samples by Western-immunoblot
analysis.[74][75][76][77] This was supported in heterozygous Mybpc3-targeted knock-in mice,[78] carrying
the human c.772G>A transition (i.e. founder mutation in Tuscany[66] These data suggest
haploinsufficiency as the main disease mechanism for heterozygous truncating mutations.[79][80] A body
of evidence exists that the mechanisms regulating the expression of mutant allele involve the nonsense-
mediated mRNA decay, the ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway
after gene transfer of mutant MYBPC3 in cardiac myocytes or in mice in vivo.[81][82][83][84][85][86] In
contrast to truncating mutations, missense mutations lead, in most of the cases (although difficult to
specifically detect), to stable mutant cMyBP-Cs that are, at least in part, incorporated into the sarcomere
and could act as poison polypeptides on the structure and/or function of the sarcomere. Homozygous or
compound heterozygous mutations are therefore likely subject to differential regulation depending on
whether they are double missense, double truncating or mixed missense/truncating mutations. The
homozygous Mybpc3-targeted knock-in mice, which genetically mimic the situation of severe neonatal
cardiomyopathy are born without phenotype and soon after birth develop systolic dysfunction followed
by (compensatory) cardiac hypertrophy.[87][88] The human c.772G>A transition results in low levels of
three different mutant Mybpc3 mRNAs and cMyBP-Cs in homozygous mice, suggesting a combination
of haploinsufficiency and polypeptide poisoning as disease mechanism in the homozygous state.[89] In
addition, the combination of external stress (such as neurohumoral stress or aging) and Mybpc3
mutations have been shown to impair the UPS in mice,[90][91] and proteasomal activities were also
depressed in patients with hypertrophic cardiomyopathy or dilated cardiomyopathy.[92]

Skinned trabeculae or cardiac myocytes obtained from human patients carrying a MYBPC3 mutation or
from heterozygous and homozygous Mybpc3-targeted knock-in mice exhibited higher myofilament Ca2+
sensitivity than controls.[93][94][95][96][97] Disease-modeling by engineered heart tissue (EHT)
technology with cardiac cells from heterozygous or homozygous Mybpc3-targeted knock-in mice
reproduced observations made in human and mouse studies displaying abbreviated contractions, greater
sensitivity to external Ca2+ and smaller inotropic responses to various drugs (isoprenaline, EMD 57033
and verapamil) compared to wild-type control EHTs.[98] Therefore, EHTs are suitable to model the
disease phenotype and recapitulate functional alterations found in mice with hypertrophic
cardiomyopathy. Another good system for modeling cardiomyopathies in the cell culture dish is the
derivation of cardiac myocytes from iPSC. Reports of human iPSC models of sarcomeric
cardiomyopathies showed cellular hypertrophy in most of the cases,[99][100][101][102] including one with
the c.2995_3010del MYBPC3 mutation that exhibited in addition to hypertrophy contractile variability in
the presence of endothelin-1.[103]

Therapy
Because of their tissue selectivity and persistent expression recombinant adeno-associated viruses (AAV)
have therapeutic potential in the treatment of inherited cardiomyopathy resulting from MYBPC3
mutations-[104] Several targeting approaches have been developed.[105][106] The most recent is genome
editing to correct a mutation by CRISPR/Cas9 technology.[107] Naturally existing as part of the
prokaryotic immune system, the CRISPR/Cas9 system has been used for correction of mutations in the
mammalian genome.[108] By inducing nicks in the double-stranded DNA and providing a template DNA
sequence, it is possible to repair mutations by homologous recombination. This approach has not yet
been evaluated for MYBPC3 mutations, but it could be used for each single or clustered mutation, and
therefore applied preferentially for frequent founder MYBPC3 mutations.

Other strategies targeting the mutant pre-mRNA by exon skipping and/or spliceosome-mediated RNA
trans-splicing (SMaRT) have been evaluated for MYBPC3. Exon skipping can be achieved using
antisense oligonucleotide (AON) masking exonic splicing enhancer sequences and therefore preventing
binding of the splicing machinery and therefore resulting in exclusion of the exon from the
mRNA.[109][110] This approach can be applied when the resulting shorter, but in-frame translated protein
maintains its function. Proof-of-concept of exon skipping was recently shown in homozygous Mybpc3-
targeted knock-in mice.[87] Systemic administration of AAV-based AONs to Mybpc3-targeted knock-in
newborn mice prevented both systolic dysfunction and left ventricular hypertrophy, at least for the
duration of the investigated period.[87] For the human MYBPC3 gene, skipping of 6 single exons or 5
double exons with specific AONs would result in shortened in-frame cMyBP-Cs, allowing the
preservation of the functionally important phosphorylation and protein interaction sites. With this
approach, about half of missense or exonic/intronic truncating mutations could be removed, including 35
mutations in exon 25. The other strategy targeting the mutant pre-mRNA is SMaRT. Hereby, two
independently transcribed molecules, the mutant pre-mRNA and the therapeutic pre-trans-splicing
molecule carrying the wild-type sequence are spliced together to give rise to a repaired full-length
mRNA.[111] Recently, the feasibility of this method was shown both in isolated cardiac myocytes and in
vivo in the heart of homozygous Mybpc3-targeted knock-in mice, although the efficiency of the process
was low and the amount of repaired protein was not sufficient to prevent the development of the cardiac
disease phenotype.[88] In principle, however, this SmART strategy is superior to exon skipping or
CRISPR/Cas9 genome editing and still attractive, because only two pre-trans-splicing molecules,
targeting the 5’ and the 3’ of MYBPC3 pre-mRNA would be sufficient to bypass all MYBPC3 mutations
associated with cardiomyopathies and therefore repair the mRNA.

AAV-mediated gene transfer of the full-length Mybpc3 (defined as “gene replacement”) dose-
dependently prevents the development of cardiac hypertrophy and dysfunction in homozygous Mybpc3-
targeted knock-in mice.[112] The dose-dependent expression of exogenous Mybpc3 was associated with
the down-regulation of endogenous mutant Mybpc3. Additional expression of a sarcomeric protein is
expected to replace partially or completely the endogenous protein level in the sarcomere, as it has been
shown in transgenic mice expressing sarcomeric proteins.[113]

Notes

References
1. GRCh38: Ensembl release 89: ENSG00000134571 (http://May2017.archive.ensembl.org/H
omo_sapiens/Gene/Summary?db=core;g=ENSG00000134571) - Ensembl, May 2017
2. GRCm38: Ensembl release 89: ENSMUSG00000002100 (http://May2017.archive.ensembl.
org/Mus_musculus/Gene/Summary?db=core;g=ENSMUSG00000002100) - Ensembl, May
2017
3. "Human PubMed Reference:" (https://www.ncbi.nlm.nih.gov/sites/entrez?db=gene&cmd=Lin
k&LinkName=gene_pubmed&from_uid=4607). National Center for Biotechnology
Information, U.S. National Library of Medicine.
4. "Mouse PubMed Reference:" (https://www.ncbi.nlm.nih.gov/sites/entrez?db=gene&cmd=Lin
k&LinkName=gene_pubmed&from_uid=17868). National Center for Biotechnology
Information, U.S. National Library of Medicine.
5. Gautel M, Zuffardi O, Freiburg A, Labeit S (May 1995). "Phosphorylation switches specific
for the cardiac isoform of myosin binding protein-C: a modulator of cardiac contraction?" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294). The EMBO Journal. 14 (9): 1952–60.
doi:10.1002/j.1460-2075.1995.tb07187.x (https://doi.org/10.1002%2Fj.1460-2075.1995.tb07
187.x). PMC 398294 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294).
PMID 7744002 (https://www.ncbi.nlm.nih.gov/pubmed/7744002).
6. Fougerousse F, Delezoide AL, Fiszman MY, Schwartz K, Beckmann JS, Carrier L (1998).
"Cardiac myosin binding protein C gene is specifically expressed in heart during murine and
human development". Circulation Research. 82 (1): 130–3. doi:10.1161/01.res.82.1.130 (htt
ps://doi.org/10.1161%2F01.res.82.1.130). PMID 9440712 (https://www.ncbi.nlm.nih.gov/pub
med/9440712).
7. Carrier L, Bonne G, Bährend E, Yu B, Richard P, Niel F, Hainque B, Cruaud C, Gary F,
Labeit S, Bouhour JB, Dubourg O, Desnos M, Hagège AA, Trent RJ, Komajda M, Fiszman
M, Schwartz K (Mar 1997). "Organization and sequence of human cardiac myosin binding
protein C gene (MYBPC3) and identification of mutations predicted to produce truncated
proteins in familial hypertrophic cardiomyopathy". Circulation Research. 80 (3): 427–34.
doi:10.1161/01.res.0000435859.24609.b3 (https://doi.org/10.1161%2F01.res.0000435859.2
4609.b3). PMID 9048664 (https://www.ncbi.nlm.nih.gov/pubmed/9048664).
8. "Protein Information - Myosin-binding protein C, cardiac-type" (http://www.heartproteome.or
g/copa/ProteinInfo.aspx?QType=Protein%20ID&QValue=Q14896). Cardiac Organellar
Protein Atlas Knowledgebase (COPaKB). NHLBI Proteomics Center at UCLA. Retrieved
29 April 2015.
9. Zong NC, Li H, Li H, Lam MP, Jimenez RC, Kim CS, Deng N, Kim AK, Choi JH, Zelaya I,
Liem D, Meyer D, Odeberg J, Fang C, Lu HJ, Xu T, Weiss J, Duan H, Uhlen M, Yates JR,
Apweiler R, Ge J, Hermjakob H, Ping P (Oct 2013). "Integration of cardiac proteome biology
and medicine by a specialized knowledgebase" (https://www.ncbi.nlm.nih.gov/pmc/articles/P
MC4076475). Circulation Research. 113 (9): 1043–53.
doi:10.1161/CIRCRESAHA.113.301151 (https://doi.org/10.1161%2FCIRCRESAHA.113.301
151). PMC 4076475 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4076475).
PMID 23965338 (https://www.ncbi.nlm.nih.gov/pubmed/23965338).
10. Bennett P, Craig R, Starr R, Offer G (Dec 1986). "The ultrastructural location of C-protein,
X-protein and H-protein in rabbit muscle". Journal of Muscle Research and Cell Motility. 7
(6): 550–67. doi:10.1007/bf01753571 (https://doi.org/10.1007%2Fbf01753571).
PMID 3543050 (https://www.ncbi.nlm.nih.gov/pubmed/3543050).
11. Offer G, Moos C, Starr R (Mar 1973). "A new protein of the thick filaments of vertebrate
skeletal myofibrils. Extractions, purification and characterization". Journal of Molecular
Biology. 74 (4): 653–76. doi:10.1016/0022-2836(73)90055-7 (https://doi.org/10.1016%2F00
22-2836%2873%2990055-7). PMID 4269687 (https://www.ncbi.nlm.nih.gov/pubmed/42696
87).
12. Freiburg A, Gautel M (Jan 1996). "A molecular map of the interactions between titin and
myosin-binding protein C. Implications for sarcomeric assembly in familial hypertrophic
cardiomyopathy". European Journal of Biochemistry / FEBS. 235 (1–2): 317–23.
doi:10.1111/j.1432-1033.1996.00317.x (https://doi.org/10.1111%2Fj.1432-1033.1996.0031
7.x). PMID 8631348 (https://www.ncbi.nlm.nih.gov/pubmed/8631348).
13. Shaffer JF, Kensler RW, Harris SP (May 2009). "The myosin-binding protein C motif binds to
F-actin in a phosphorylation-sensitive manner" (https://www.ncbi.nlm.nih.gov/pmc/articles/P
MC2673300). The Journal of Biological Chemistry. 284 (18): 12318–27.
doi:10.1074/jbc.M808850200 (https://doi.org/10.1074%2Fjbc.M808850200). PMC 2673300
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2673300). PMID 19269976 (https://www.ncb
i.nlm.nih.gov/pubmed/19269976).
14. Winegrad S (May 1999). "Cardiac myosin binding protein C". Circulation Research. 84 (10):
1117–26. doi:10.1161/01.res.84.10.1117 (https://doi.org/10.1161%2F01.res.84.10.1117).
PMID 10347086 (https://www.ncbi.nlm.nih.gov/pubmed/10347086).
15. Koretz JF (Sep 1979). "Effects of C-protein on synthetic myosin filament structure" (https://w
ww.ncbi.nlm.nih.gov/pmc/articles/PMC1328598). Biophysical Journal. 27 (3): 433–46.
doi:10.1016/S0006-3495(79)85227-3 (https://doi.org/10.1016%2FS0006-3495%2879%2985
227-3). PMC 1328598 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1328598).
PMID 263692 (https://www.ncbi.nlm.nih.gov/pubmed/263692).
16. Colson BA, Bekyarova T, Fitzsimons DP, Irving TC, Moss RL (Mar 2007). "Radial
displacement of myosin cross-bridges in mouse myocardium due to ablation of myosin
binding protein-C" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1892277). Journal of
Molecular Biology. 367 (1): 36–41. doi:10.1016/j.jmb.2006.12.063 (https://doi.org/10.1016%
2Fj.jmb.2006.12.063). PMC 1892277 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC18922
77). PMID 17254601 (https://www.ncbi.nlm.nih.gov/pubmed/17254601).
17. Harris SP, Bartley CR, Hacker TA, McDonald KS, Douglas PS, Greaser ML, Powers PA,
Moss RL (Mar 2002). "Hypertrophic cardiomyopathy in cardiac myosin binding protein-C
knockout mice". Circulation Research. 90 (5): 594–601.
doi:10.1161/01.res.0000012222.70819.64 (https://doi.org/10.1161%2F01.res.0000012222.7
0819.64). PMID 11909824 (https://www.ncbi.nlm.nih.gov/pubmed/11909824).
18. Carrier L, Knöll R, Vignier N, Keller DI, Bausero P, Prudhon B, Isnard R, Ambroisine ML,
Fiszman M, Ross J, Schwartz K, Chien KR (Aug 2004). "Asymmetric septal hypertrophy in
heterozygous cMyBP-C null mice". Cardiovascular Research. 63 (2): 293–304.
doi:10.1016/j.cardiores.2004.04.009 (https://doi.org/10.1016%2Fj.cardiores.2004.04.009).
PMID 15249187 (https://www.ncbi.nlm.nih.gov/pubmed/15249187).
19. Cazorla O, Szilagyi S, Vignier N, Salazar G, Krämer E, Vassort G, Carrier L, Lacampagne A
(Feb 2006). "Length and protein kinase A modulations of myocytes in cardiac myosin
binding protein C-deficient mice". Cardiovascular Research. 69 (2): 370–80.
doi:10.1016/j.cardiores.2005.11.009 (https://doi.org/10.1016%2Fj.cardiores.2005.11.009).
PMID 16380103 (https://www.ncbi.nlm.nih.gov/pubmed/16380103).
20. Brickson S, Fitzsimons DP, Pereira L, Hacker T, Valdivia H, Moss RL (Apr 2007). "In vivo left
ventricular functional capacity is compromised in cMyBP-C null mice". American Journal of
Physiology. Heart and Circulatory Physiology. 292 (4): H1747–54.
doi:10.1152/ajpheart.01037.2006 (https://doi.org/10.1152%2Fajpheart.01037.2006).
PMID 17122190 (https://www.ncbi.nlm.nih.gov/pubmed/17122190).
21. Luther PK, Bennett PM, Knupp C, Craig R, Padrón R, Harris SP, Patel J, Moss RL (Dec
2008). "Understanding the organisation and role of myosin binding protein C in normal
striated muscle by comparison with MyBP-C knockout cardiac muscle" (https://www.ncbi.nl
m.nih.gov/pmc/articles/PMC2593797). Journal of Molecular Biology. 384 (1): 60–72.
doi:10.1016/j.jmb.2008.09.013 (https://doi.org/10.1016%2Fj.jmb.2008.09.013).
PMC 2593797 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2593797). PMID 18817784
(https://www.ncbi.nlm.nih.gov/pubmed/18817784).
22. Korte FS, McDonald KS, Harris SP, Moss RL (Oct 2003). "Loaded shortening, power output,
and rate of force redevelopment are increased with knockout of cardiac myosin binding
protein-C". Circulation Research. 93 (8): 752–8.
doi:10.1161/01.RES.0000096363.85588.9A (https://doi.org/10.1161%2F01.RES.000009636
3.85588.9A). PMID 14500336 (https://www.ncbi.nlm.nih.gov/pubmed/14500336).
23. Palmer BM, Georgakopoulos D, Janssen PM, Wang Y, Alpert NR, Belardi DF, Harris SP,
Moss RL, Burgon PG, Seidman CE, Seidman JG, Maughan DW, Kass DA (May 2004).
"Role of cardiac myosin binding protein C in sustaining left ventricular systolic stiffening".
Circulation Research. 94 (9): 1249–55. doi:10.1161/01.RES.0000126898.95550.31 (https://
doi.org/10.1161%2F01.RES.0000126898.95550.31). PMID 15059932 (https://www.ncbi.nl
m.nih.gov/pubmed/15059932).
24. Stelzer JE, Fitzsimons DP, Moss RL (Jun 2006). "Ablation of myosin-binding protein-C
accelerates force development in mouse myocardium" (https://www.ncbi.nlm.nih.gov/pmc/ar
ticles/PMC1459529). Biophysical Journal. 90 (11): 4119–27.
doi:10.1529/biophysj.105.078147 (https://doi.org/10.1529%2Fbiophysj.105.078147).
PMC 1459529 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1459529). PMID 16513777
(https://www.ncbi.nlm.nih.gov/pubmed/16513777).
25. Gruen M, Gautel M (Feb 1999). "Mutations in beta-myosin S2 that cause familial
hypertrophic cardiomyopathy (FHC) abolish the interaction with the regulatory domain of
myosin-binding protein-C". Journal of Molecular Biology. 286 (3): 933–49.
doi:10.1006/jmbi.1998.2522 (https://doi.org/10.1006%2Fjmbi.1998.2522). PMID 10024460
(https://www.ncbi.nlm.nih.gov/pubmed/10024460).
26. Kunst G, Kress KR, Gruen M, Uttenweiler D, Gautel M, Fink RH (2000). "Myosin binding
protein C, a phosphorylation-dependent force regulator in muscle that controls the
attachment of myosin heads by its interaction with myosin S2". Circulation Research. 86 (1):
51–8. doi:10.1161/01.res.86.1.51 (https://doi.org/10.1161%2F01.res.86.1.51).
PMID 10625305 (https://www.ncbi.nlm.nih.gov/pubmed/10625305).
27. Harris SP, Rostkova E, Gautel M, Moss RL (Oct 2004). "Binding of myosin binding protein-C
to myosin subfragment S2 affects contractility independent of a tether mechanism".
Circulation Research. 95 (9): 930–6. doi:10.1161/01.RES.0000147312.02673.56 (https://do
i.org/10.1161%2F01.RES.0000147312.02673.56). PMID 15472117 (https://www.ncbi.nlm.ni
h.gov/pubmed/15472117).
28. Ababou A, Gautel M, Pfuhl M (Mar 2007). "Dissecting the N-terminal myosin binding site of
human cardiac myosin-binding protein C. Structure and myosin binding of domain C2". The
Journal of Biological Chemistry. 282 (12): 9204–15. doi:10.1074/jbc.M610899200 (https://do
i.org/10.1074%2Fjbc.M610899200). PMID 17192269 (https://www.ncbi.nlm.nih.gov/pubme
d/17192269).
29. Pohlmann L, Kröger I, Vignier N, Schlossarek S, Krämer E, Coirault C, Sultan KR, El-
Armouche A, Winegrad S, Eschenhagen T, Carrier L (Oct 2007). "Cardiac myosin-binding
protein C is required for complete relaxation in intact myocytes". Circulation Research. 101
(9): 928–38. doi:10.1161/CIRCRESAHA.107.158774 (https://doi.org/10.1161%2FCIRCRES
AHA.107.158774). PMID 17823372 (https://www.ncbi.nlm.nih.gov/pubmed/17823372).
30. Moss RL, Fitzsimons DP, Ralphe JC (Jan 2015). "Cardiac MyBP-C regulates the rate and
force of contraction in mammalian myocardium" (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC4283578). Circulation Research. 116 (1): 183–92.
doi:10.1161/CIRCRESAHA.116.300561 (https://doi.org/10.1161%2FCIRCRESAHA.116.300
561). PMC 4283578 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4283578).
PMID 25552695 (https://www.ncbi.nlm.nih.gov/pubmed/25552695).
31. Hartzell HC, Titus L (Feb 1982). "Effects of cholinergic and adrenergic agonists on
phosphorylation of a 165,000-dalton myofibrillar protein in intact cardiac muscle". The
Journal of Biological Chemistry. 257 (4): 2111–20. PMID 6276407 (https://www.ncbi.nlm.ni
h.gov/pubmed/6276407).
32. Hartzell HC, Glass DB (Dec 1984). "Phosphorylation of purified cardiac muscle C-protein by
purified cAMP-dependent and endogenous Ca2+-calmodulin-dependent protein kinases".
The Journal of Biological Chemistry. 259 (24): 15587–96. PMID 6549009 (https://www.ncbi.
nlm.nih.gov/pubmed/6549009).
33. Gautel M, Zuffardi O, Freiburg A, Labeit S (May 1995). "Phosphorylation switches specific
for the cardiac isoform of myosin binding protein-C: a modulator of cardiac contraction?" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294). The EMBO Journal. 14 (9): 1952–60.
doi:10.1002/j.1460-2075.1995.tb07187.x (https://doi.org/10.1002%2Fj.1460-2075.1995.tb07
187.x). PMC 398294 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294).
PMID 7744002 (https://www.ncbi.nlm.nih.gov/pubmed/7744002).
34. Mohamed AS, Dignam JD, Schlender KK (Oct 1998). "Cardiac myosin-binding protein C
(MyBP-C): identification of protein kinase A and protein kinase C phosphorylation sites".
Archives of Biochemistry and Biophysics. 358 (2): 313–9. doi:10.1006/abbi.1998.0857 (http
s://doi.org/10.1006%2Fabbi.1998.0857). PMID 9784245 (https://www.ncbi.nlm.nih.gov/pub
med/9784245).
35. McClellan G, Kulikovskaya I, Winegrad S (Aug 2001). "Changes in cardiac contractility
related to calcium-mediated changes in phosphorylation of myosin-binding protein C" (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC1301577). Biophysical Journal. 81 (2): 1083–92.
doi:10.1016/S0006-3495(01)75765-7 (https://doi.org/10.1016%2FS0006-3495%2801%2975
765-7). PMC 1301577 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1301577).
PMID 11463649 (https://www.ncbi.nlm.nih.gov/pubmed/11463649).
36. Sadayappan S, Gulick J, Osinska H, Barefield D, Cuello F, Avkiran M, Lasko VM, Lorenz
JN, Maillet M, Martin JL, Brown JH, Bers DM, Molkentin JD, James J, Robbins J (Jul 2011).
"A critical function for Ser-282 in cardiac Myosin binding protein-C phosphorylation and
cardiac function" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3132348). Circulation
Research. 109 (2): 141–50. doi:10.1161/CIRCRESAHA.111.242560 (https://doi.org/10.116
1%2FCIRCRESAHA.111.242560). PMC 3132348 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC3132348). PMID 21597010 (https://www.ncbi.nlm.nih.gov/pubmed/21597010).
37. Cuello F, Bardswell SC, Haworth RS, Ehler E, Sadayappan S, Kentish JC, Avkiran M (Feb
2011). "Novel role for p90 ribosomal S6 kinase in the regulation of cardiac myofilament
phosphorylation" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3037642). The Journal of
Biological Chemistry. 286 (7): 5300–10. doi:10.1074/jbc.M110.202713 (https://doi.org/10.10
74%2Fjbc.M110.202713). PMC 3037642 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC30
37642). PMID 21148481 (https://www.ncbi.nlm.nih.gov/pubmed/21148481).
38. Bardswell SC, Cuello F, Rowland AJ, Sadayappan S, Robbins J, Gautel M, Walker JW,
Kentish JC, Avkiran M (Feb 2010). "Distinct sarcomeric substrates are responsible for
protein kinase D-mediated regulation of cardiac myofilament Ca2+ sensitivity and cross-
bridge cycling" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2820795). The Journal of
Biological Chemistry. 285 (8): 5674–82. doi:10.1074/jbc.M109.066456 (https://doi.org/10.10
74%2Fjbc.M109.066456). PMC 2820795 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC28
20795). PMID 20018870 (https://www.ncbi.nlm.nih.gov/pubmed/20018870).
39. Dirkx E, Cazorla O, Schwenk RW, Lorenzen-Schmidt I, Sadayappan S, Van Lint J, Carrier
L, van Eys GJ, Glatz JF, Luiken JJ (Aug 2012). "Protein kinase D increases maximal Ca2+-
activated tension of cardiomyocyte contraction by phosphorylation of cMyBP-C-Ser315" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC6734090). American Journal of Physiology.
Heart and Circulatory Physiology. 303 (3): H323–31. doi:10.1152/ajpheart.00749.2011 (http
s://doi.org/10.1152%2Fajpheart.00749.2011). PMC 6734090 (https://www.ncbi.nlm.nih.gov/
pmc/articles/PMC6734090). PMID 22636676 (https://www.ncbi.nlm.nih.gov/pubmed/226366
76).
40. Kuster DW, Sequeira V, Najafi A, Boontje NM, Wijnker PJ, Witjas-Paalberends ER, Marston
SB, Dos Remedios CG, Carrier L, Demmers JA, Redwood C, Sadayappan S, van der
Velden J (Feb 2013). "GSK3β phosphorylates newly identified site in the proline-alanine-rich
region of cardiac myosin-binding protein C and alters cross-bridge cycling kinetics in
human: short communication" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3595322).
Circulation Research. 112 (4): 633–9. doi:10.1161/CIRCRESAHA.112.275602 (https://doi.or
g/10.1161%2FCIRCRESAHA.112.275602). PMC 3595322 (https://www.ncbi.nlm.nih.gov/p
mc/articles/PMC3595322). PMID 23277198 (https://www.ncbi.nlm.nih.gov/pubmed/2327719
8).
41. Govindan S, Sarkey J, Ji X, Sundaresan NR, Gupta MP, de Tombe PP, Sadayappan S (May
2012). "Pathogenic properties of the N-terminal region of cardiac myosin binding protein-C
in vitro" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3368277). Journal of Muscle
Research and Cell Motility. 33 (1): 17–30. doi:10.1007/s10974-012-9292-y (https://doi.org/1
0.1007%2Fs10974-012-9292-y). PMC 3368277 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC3368277). PMID 22527638 (https://www.ncbi.nlm.nih.gov/pubmed/22527638).
42. Witayavanitkul N, Ait Mou Y, Kuster DW, Khairallah RJ, Sarkey J, Govindan S, Chen X, Ge
Y, Rajan S, Wieczorek DF, Irving T, Westfall MV, de Tombe PP, Sadayappan S (Mar 2014).
"Myocardial infarction-induced N-terminal fragment of cardiac myosin-binding protein C
(cMyBP-C) impairs myofilament function in human myocardium" (https://www.ncbi.nlm.nih.g
ov/pmc/articles/PMC3979389). The Journal of Biological Chemistry. 289 (13): 8818–27.
doi:10.1074/jbc.M113.541128 (https://doi.org/10.1074%2Fjbc.M113.541128).
PMC 3979389 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3979389). PMID 24509847
(https://www.ncbi.nlm.nih.gov/pubmed/24509847).
43. El-Armouche A, Pohlmann L, Schlossarek S, Starbatty J, Yeh YH, Nattel S, Dobrev D,
Eschenhagen T, Carrier L (Aug 2007). "Decreased phosphorylation levels of cardiac
myosin-binding protein-C in human and experimental heart failure". Journal of Molecular
and Cellular Cardiology. 43 (2): 223–9. doi:10.1016/j.yjmcc.2007.05.003 (https://doi.org/10.
1016%2Fj.yjmcc.2007.05.003). PMID 17560599 (https://www.ncbi.nlm.nih.gov/pubmed/175
60599).
44. Copeland O, Sadayappan S, Messer AE, Steinen GJ, van der Velden J, Marston SB (Dec
2010). "Analysis of cardiac myosin binding protein-C phosphorylation in human heart
muscle" (https://zenodo.org/record/3423484). Journal of Molecular and Cellular Cardiology.
49 (6): 1003–11. doi:10.1016/j.yjmcc.2010.09.007 (https://doi.org/10.1016%2Fj.yjmcc.2010.
09.007). PMID 20850451 (https://www.ncbi.nlm.nih.gov/pubmed/20850451).
45. Ge Y, Rybakova IN, Xu Q, Moss RL (Aug 2009). "Top-down high-resolution mass
spectrometry of cardiac myosin binding protein C revealed that truncation alters protein
phosphorylation state" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2722289).
Proceedings of the National Academy of Sciences of the United States of America. 106
(31): 12658–63. doi:10.1073/pnas.0813369106 (https://doi.org/10.1073%2Fpnas.08133691
06). PMC 2722289 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2722289).
PMID 19541641 (https://www.ncbi.nlm.nih.gov/pubmed/19541641).
46. Fert-Bober J, Sokolove J (Aug 2014). "Proteomics of citrullination in cardiovascular
disease". Proteomics: Clinical Applications. 8 (7–8): 522–33. doi:10.1002/prca.201400013
(https://doi.org/10.1002%2Fprca.201400013). PMID 24946285 (https://www.ncbi.nlm.nih.go
v/pubmed/24946285).
47. Brennan JP, Miller JI, Fuller W, Wait R, Begum S, Dunn MJ, Eaton P (Feb 2006). "The utility
of N,N-biotinyl glutathione disulfide in the study of protein S-glutathiolation". Molecular &
Cellular Proteomics. 5 (2): 215–25. doi:10.1074/mcp.M500212-MCP200 (https://doi.org/10.1
074%2Fmcp.M500212-MCP200). PMID 16223748 (https://www.ncbi.nlm.nih.gov/pubmed/1
6223748).
48. Lovelock JD, Monasky MM, Jeong EM, Lardin HA, Liu H, Patel BG, Taglieri DM, Gu L,
Kumar P, Pokhrel N, Zeng D, Belardinelli L, Sorescu D, Solaro RJ, Dudley SC (Mar 2012).
"Ranolazine improves cardiac diastolic dysfunction through modulation of myofilament
calcium sensitivity" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3314887). Circulation
Research. 110 (6): 841–50. doi:10.1161/CIRCRESAHA.111.258251 (https://doi.org/10.116
1%2FCIRCRESAHA.111.258251). PMC 3314887 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC3314887). PMID 22343711 (https://www.ncbi.nlm.nih.gov/pubmed/22343711).
49. Jeong EM, Monasky MM, Gu L, Taglieri DM, Patel BG, Liu H, Wang Q, Greener I, Dudley
SC, Solaro RJ (Mar 2013). "Tetrahydrobiopterin improves diastolic dysfunction by reversing
changes in myofilament properties" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC366658
5). Journal of Molecular and Cellular Cardiology. 56: 44–54.
doi:10.1016/j.yjmcc.2012.12.003 (https://doi.org/10.1016%2Fj.yjmcc.2012.12.003).
PMC 3666585 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3666585). PMID 23247392
(https://www.ncbi.nlm.nih.gov/pubmed/23247392).
50. Patel BG, Wilder T, Solaro RJ (2013). "Novel control of cardiac myofilament response to
calcium by S-glutathionylation at specific sites of myosin binding protein C" (https://www.ncb
i.nlm.nih.gov/pmc/articles/PMC3834529). Frontiers in Physiology. 4: 336.
doi:10.3389/fphys.2013.00336 (https://doi.org/10.3389%2Ffphys.2013.00336).
PMC 3834529 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3834529). PMID 24312057
(https://www.ncbi.nlm.nih.gov/pubmed/24312057).
51. Kohr MJ, Aponte AM, Sun J, Wang G, Murphy E, Gucek M, Steenbergen C (Apr 2011).
"Characterization of potential S-nitrosylation sites in the myocardium" (https://www.ncbi.nlm.
nih.gov/pmc/articles/PMC3075037). American Journal of Physiology. Heart and Circulatory
Physiology. 300 (4): H1327–35. doi:10.1152/ajpheart.00997.2010 (https://doi.org/10.1152%
2Fajpheart.00997.2010). PMC 3075037 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC307
5037). PMID 21278135 (https://www.ncbi.nlm.nih.gov/pubmed/21278135).
52. Aryal B, Jeong J, Rao VA (Feb 2014). "Doxorubicin-induced carbonylation and degradation
of cardiac myosin binding protein C promote cardiotoxicity" (https://www.ncbi.nlm.nih.gov/p
mc/articles/PMC3918758). Proceedings of the National Academy of Sciences of the United
States of America. 111 (5): 2011–6. doi:10.1073/pnas.1321783111 (https://doi.org/10.107
3%2Fpnas.1321783111). PMC 3918758 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC39
18758). PMID 24449919 (https://www.ncbi.nlm.nih.gov/pubmed/24449919).
53. Gautel M, Zuffardi O, Freiburg A, Labeit S (May 1995). "Phosphorylation switches specific
for the cardiac isoform of myosin binding protein-C: a modulator of cardiac contraction?" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294). The EMBO Journal. 14 (9): 1952–60.
doi:10.1002/j.1460-2075.1995.tb07187.x (https://doi.org/10.1002%2Fj.1460-2075.1995.tb07
187.x). PMC 398294 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294).
PMID 7744002 (https://www.ncbi.nlm.nih.gov/pubmed/7744002).
54. Carrier L, Hengstenberg C, Beckmann JS, Guicheney P, Dufour C, Bercovici J, Dausse E,
Berebbi-Bertrand I, Wisnewsky C, Pulvenis D (Jul 1993). "Mapping of a novel gene for
familial hypertrophic cardiomyopathy to chromosome 11". Nature Genetics. 4 (3): 311–3.
doi:10.1038/ng0793-311 (https://doi.org/10.1038%2Fng0793-311). PMID 8358441 (https://w
ww.ncbi.nlm.nih.gov/pubmed/8358441).
55. Bonne G, Carrier L, Bercovici J, Cruaud C, Richard P, Hainque B, Gautel M, Labeit S,
James M, Beckmann J, Weissenbach J, Vosberg HP, Fiszman M, Komajda M, Schwartz K
(Dec 1995). "Cardiac myosin binding protein-C gene splice acceptor site mutation is
associated with familial hypertrophic cardiomyopathy". Nature Genetics. 11 (4): 438–40.
doi:10.1038/ng1295-438 (https://doi.org/10.1038%2Fng1295-438). PMID 7493026 (https://w
ww.ncbi.nlm.nih.gov/pubmed/7493026).
56. Watkins H, Conner D, Thierfelder L, Jarcho JA, MacRae C, McKenna WJ, Maron BJ,
Seidman JG, Seidman CE (Dec 1995). "Mutations in the cardiac myosin binding protein-C
gene on chromosome 11 cause familial hypertrophic cardiomyopathy". Nature Genetics. 11
(4): 434–7. doi:10.1038/ng1295-434 (https://doi.org/10.1038%2Fng1295-434).
PMID 7493025 (https://www.ncbi.nlm.nih.gov/pubmed/7493025).
57. Harris SP, Lyons RG, Bezold KL (Mar 2011). "In the thick of it: HCM-causing mutations in
myosin binding proteins of the thick filament" (https://www.ncbi.nlm.nih.gov/pmc/articles/PM
C3076008). Circulation Research. 108 (6): 751–64. doi:10.1161/CIRCRESAHA.110.231670
(https://doi.org/10.1161%2FCIRCRESAHA.110.231670). PMC 3076008 (https://www.ncbi.nl
m.nih.gov/pmc/articles/PMC3076008). PMID 21415409 (https://www.ncbi.nlm.nih.gov/pubm
ed/21415409).
58. Behrens-Gawlik V, Mearini G, Gedicke-Hornung C, Richard P, Carrier L (Feb 2014).
"MYBPC3 in hypertrophic cardiomyopathy: from mutation identification to RNA-based
correction". Pflügers Archiv. 466 (2): 215–23. doi:10.1007/s00424-013-1409-7 (https://doi.or
g/10.1007%2Fs00424-013-1409-7). PMID 24337823 (https://www.ncbi.nlm.nih.gov/pubme
d/24337823).
59. Haas J, Frese KS, Peil B, Kloos W, Keller A, Nietsch R, Feng Z, Müller S, Kayvanpour E,
Vogel B, Sedaghat-Hamedani F, Lim WK, Zhao X, Fradkin D, Köhler D, Fischer S, Franke J,
Marquart S, Barb I, Li DT, Amr A, Ehlermann P, Mereles D, Weis T, Hassel S, Kremer A,
King V, Wirsz E, Isnard R, Komajda M, Serio A, Grasso M, Syrris P, Wicks E, Plagnol V,
Lopes L, Gadgaard T, Eiskjær H, Jørgensen M, Garcia-Giustiniani D, Ortiz-Genga M,
Crespo-Leiro MG, Deprez RH, Christiaans I, van Rijsingen IA, Wilde AA, Waldenstrom A,
Bolognesi M, Bellazzi R, Mörner S, Bermejo JL, Monserrat L, Villard E, Mogensen J, Pinto
YM, Charron P, Elliott P, Arbustini E, Katus HA, Meder B (May 2015). "Atlas of the clinical
genetics of human dilated cardiomyopathy". European Heart Journal. 36 (18): 1123–35.
doi:10.1093/eurheartj/ehu301 (https://doi.org/10.1093%2Feurheartj%2Fehu301).
PMID 25163546 (https://www.ncbi.nlm.nih.gov/pubmed/25163546).
60. Probst S, Oechslin E, Schuler P, Greutmann M, Boyé P, Knirsch W, Berger F, Thierfelder L,
Jenni R, Klaassen S (Aug 2011). "Sarcomere gene mutations in isolated left ventricular
noncompaction cardiomyopathy do not predict clinical phenotype". Circulation:
Cardiovascular Genetics. 4 (4): 367–74. doi:10.1161/CIRCGENETICS.110.959270 (https://
doi.org/10.1161%2FCIRCGENETICS.110.959270). PMID 21551322 (https://www.ncbi.nlm.
nih.gov/pubmed/21551322).
61. Dhandapany PS, Sadayappan S, Xue Y, Powell GT, Rani DS, Nallari P, Rai TS, Khullar M,
Soares P, Bahl A, Tharkan JM, Vaideeswar P, Rathinavel A, Narasimhan C, Ayapati DR,
Ayub Q, Mehdi SQ, Oppenheimer S, Richards MB, Price AL, Patterson N, Reich D, Singh L,
Tyler-Smith C, Thangaraj K (Feb 2009). "A common MYBPC3 (cardiac myosin binding
protein C) variant associated with cardiomyopathies in South Asia" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC2697598). Nature Genetics. 41 (2): 187–91. doi:10.1038/ng.309 (htt
ps://doi.org/10.1038%2Fng.309). PMC 2697598 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC2697598). PMID 19151713 (https://www.ncbi.nlm.nih.gov/pubmed/19151713).
62. Adalsteinsdottir B, Teekakirikul P, Maron BJ, Burke MA, Gudbjartsson DF, Holm H,
Stefansson K, DePalma SR, Mazaika E, McDonough B, Danielsen R, Seidman JG,
Seidman CE, Gunnarsson GT (Sep 2014). "Nationwide study on hypertrophic
cardiomyopathy in Iceland: evidence of a MYBPC3 founder mutation". Circulation. 130 (14):
1158–67. doi:10.1161/CIRCULATIONAHA.114.011207 (https://doi.org/10.1161%2FCIRCUL
ATIONAHA.114.011207). PMID 25078086 (https://www.ncbi.nlm.nih.gov/pubmed/2507808
6).
63. Calore C, De Bortoli M, Romualdi C, Lorenzon A, Angelini A, Basso C, Thiene G, Iliceto S,
Rampazzo A, Melacini P (May 2015). "A founder MYBPC3 mutation results in HCM with a
high risk of sudden death after the fourth decade of life". Journal of Medical Genetics. 52
(5): 338–47. doi:10.1136/jmedgenet-2014-102923 (https://doi.org/10.1136%2Fjmedgenet-2
014-102923). PMID 25740977 (https://www.ncbi.nlm.nih.gov/pubmed/25740977).
64. Christiaans I, Nannenberg EA, Dooijes D, Jongbloed RJ, Michels M, Postema PG, Majoor-
Krakauer D, van den Wijngaard A, Mannens MM, van Tintelen JP, van Langen IM, Wilde AA
(May 2010). "Founder mutations in hypertrophic cardiomyopathy patients in the
Netherlands" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2871745). Netherlands Heart
Journal. 18 (5): 248–54. doi:10.1007/bf03091771 (https://doi.org/10.1007%2Fbf03091771).
PMC 2871745 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2871745). PMID 20505798
(https://www.ncbi.nlm.nih.gov/pubmed/20505798).
65. Kubo T, Kitaoka H, Okawa M, Matsumura Y, Hitomi N, Yamasaki N, Furuno T, Takata J,
Nishinaga M, Kimura A, Doi YL (Nov 2005). "Lifelong left ventricular remodeling of
hypertrophic cardiomyopathy caused by a founder frameshift deletion mutation in the
cardiac Myosin-binding protein C gene among Japanese". Journal of the American College
of Cardiology. 46 (9): 1737–43. doi:10.1016/j.jacc.2005.05.087 (https://doi.org/10.1016%2F
j.jacc.2005.05.087). PMID 16256878 (https://www.ncbi.nlm.nih.gov/pubmed/16256878).
66. Girolami F, Olivotto I, Passerini I, Zachara E, Nistri S, Re F, Fantini S, Baldini K, Torricelli F,
Cecchi F (Aug 2006). "A molecular screening strategy based on beta-myosin heavy chain,
cardiac myosin binding protein C and troponin T genes in Italian patients with hypertrophic
cardiomyopathy". Journal of Cardiovascular Medicine. 7 (8): 601–7.
doi:10.2459/01.JCM.0000237908.26377.d6 (https://doi.org/10.2459%2F01.JCM.000023790
8.26377.d6). PMID 16858239 (https://www.ncbi.nlm.nih.gov/pubmed/16858239).
67. Teirlinck CH, Senni F, Malti RE, Majoor-Krakauer D, Fellmann F, Millat G, André-Fouët X,
Pernot F, Stumpf M, Boutarin J, Bouvagnet P (2012). "A human MYBPC3 mutation
appearing about 10 centuries ago results in a hypertrophic cardiomyopathy with delayed
onset, moderate evolution but with a risk of sudden death" (https://www.ncbi.nlm.nih.gov/pm
c/articles/PMC3549277). BMC Medical Genetics. 13: 105. doi:10.1186/1471-2350-13-105
(https://doi.org/10.1186%2F1471-2350-13-105). PMC 3549277 (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC3549277). PMID 23140321 (https://www.ncbi.nlm.nih.gov/pubmed/2314
0321).
68. Jääskeläinen P, Miettinen R, Kärkkäinen P, Toivonen L, Laakso M, Kuusisto J (2004).
"Genetics of hypertrophic cardiomyopathy in eastern Finland: few founder mutations with
benign or intermediary phenotypes". Annals of Medicine. 36 (1): 23–32.
doi:10.1080/07853890310017161 (https://doi.org/10.1080%2F07853890310017161).
PMID 15000344 (https://www.ncbi.nlm.nih.gov/pubmed/15000344).
69. Richard P, Charron P, Carrier L, Ledeuil C, Cheav T, Pichereau C, Benaiche A, Isnard R,
Dubourg O, Burban M, Gueffet JP, Millaire A, Desnos M, Schwartz K, Hainque B, Komajda
M (May 2003). "Hypertrophic cardiomyopathy: distribution of disease genes, spectrum of
mutations, and implications for a molecular diagnosis strategy". Circulation. 107 (17): 2227–
32. doi:10.1161/01.CIR.0000066323.15244.54 (https://doi.org/10.1161%2F01.CIR.0000066
323.15244.54). PMID 12707239 (https://www.ncbi.nlm.nih.gov/pubmed/12707239).
70. Morita H, Rehm HL, Menesses A, McDonough B, Roberts AE, Kucherlapati R, Towbin JA,
Seidman JG, Seidman CE (May 2008). "Shared genetic causes of cardiac hypertrophy in
children and adults" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2752150). The New
England Journal of Medicine. 358 (18): 1899–908. doi:10.1056/NEJMoa075463 (https://doi.
org/10.1056%2FNEJMoa075463). PMC 2752150 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC2752150). PMID 18403758 (https://www.ncbi.nlm.nih.gov/pubmed/18403758).
71. Wessels MW, Herkert JC, Frohn-Mulder IM, Dalinghaus M, van den Wijngaard A, de Krijger
RR, Michels M, de Coo IF, Hoedemaekers YM, Dooijes D (Oct 2014). "Compound
heterozygous or homozygous truncating MYBPC3 mutations cause lethal cardiomyopathy
with features of noncompaction and septal defects" (https://www.ncbi.nlm.nih.gov/pmc/articl
es/PMC4463499). European Journal of Human Genetics. 23 (7): 922–8.
doi:10.1038/ejhg.2014.211 (https://doi.org/10.1038%2Fejhg.2014.211). PMC 4463499 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC4463499). PMID 25335496 (https://www.ncbi.nl
m.nih.gov/pubmed/25335496).
72. Duncker DJ, Bakkers J, Brundel BJ, Robbins J, Tardiff JC, Carrier L (Apr 2015). "Animal
and in silico models for the study of sarcomeric cardiomyopathies" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC4375391). Cardiovascular Research. 105 (4): 439–48.
doi:10.1093/cvr/cvv006 (https://doi.org/10.1093%2Fcvr%2Fcvv006). PMC 4375391 (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC4375391). PMID 25600962 (https://www.ncbi.nlm.ni
h.gov/pubmed/25600962).
73. Eschenhagen T, Mummery C, Knollmann BC (Apr 2015). "Modelling sarcomeric
cardiomyopathies in the dish: from human heart samples to iPSC cardiomyocytes" (https://w
ww.ncbi.nlm.nih.gov/pmc/articles/PMC4349163). Cardiovascular Research. 105 (4): 424–
38. doi:10.1093/cvr/cvv017 (https://doi.org/10.1093%2Fcvr%2Fcvv017). PMC 4349163 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC4349163). PMID 25618410 (https://www.ncbi.nl
m.nih.gov/pubmed/25618410).
74. Rottbauer W, Gautel M, Zehelein J, Labeit S, Franz WM, Fischer C, Vollrath B, Mall G, Dietz
R, Kübler W, Katus HA (Jul 1997). "Novel splice donor site mutation in the cardiac myosin-
binding protein-C gene in familial hypertrophic cardiomyopathy. Characterization Of cardiac
transcript and protein" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC508212). The Journal
of Clinical Investigation. 100 (2): 475–82. doi:10.1172/JCI119555 (https://doi.org/10.1172%
2FJCI119555). PMC 508212 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC508212).
PMID 9218526 (https://www.ncbi.nlm.nih.gov/pubmed/9218526).
75. Moolman JA, Reith S, Uhl K, Bailey S, Gautel M, Jeschke B, Fischer C, Ochs J, McKenna
WJ, Klues H, Vosberg HP (Mar 2000). "A newly created splice donor site in exon 25 of the
MyBP-C gene is responsible for inherited hypertrophic cardiomyopathy with incomplete
disease penetrance". Circulation. 101 (12): 1396–402. doi:10.1161/01.cir.101.12.1396 (http
s://doi.org/10.1161%2F01.cir.101.12.1396). PMID 10736283 (https://www.ncbi.nlm.nih.gov/
pubmed/10736283).
76. Marston S, Copeland O, Jacques A, Livesey K, Tsang V, McKenna WJ, Jalilzadeh S,
Carballo S, Redwood C, Watkins H (Jul 2009). "Evidence from human myectomy samples
that MYBPC3 mutations cause hypertrophic cardiomyopathy through haploinsufficiency".
Circulation Research. 105 (3): 219–22. doi:10.1161/CIRCRESAHA.109.202440 (https://doi.
org/10.1161%2FCIRCRESAHA.109.202440). PMID 19574547 (https://www.ncbi.nlm.nih.go
v/pubmed/19574547).
77. van Dijk SJ, Dooijes D, dos Remedios C, Michels M, Lamers JM, Winegrad S, Schlossarek
S, Carrier L, ten Cate FJ, Stienen GJ, van der Velden J (Mar 2009). "Cardiac myosin-
binding protein C mutations and hypertrophic cardiomyopathy: haploinsufficiency, deranged
phosphorylation, and cardiomyocyte dysfunction". Circulation. 119 (11): 1473–83.
doi:10.1161/CIRCULATIONAHA.108.838672 (https://doi.org/10.1161%2FCIRCULATIONAH
A.108.838672). PMID 19273718 (https://www.ncbi.nlm.nih.gov/pubmed/19273718).
78. Vignier N, Schlossarek S, Fraysse B, Mearini G, Krämer E, Pointu H, Mougenot N, Guiard
J, Reimer R, Hohenberg H, Schwartz K, Vernet M, Eschenhagen T, Carrier L (Jul 2009).
"Nonsense-mediated mRNA decay and ubiquitin-proteasome system regulate cardiac
myosin-binding protein C mutant levels in cardiomyopathic mice". Circulation Research. 105
(3): 239–48. doi:10.1161/CIRCRESAHA.109.201251 (https://doi.org/10.1161%2FCIRCRES
AHA.109.201251). PMID 19590044 (https://www.ncbi.nlm.nih.gov/pubmed/19590044).
79. Marston S, Copeland O, Gehmlich K, Schlossarek S, Carrier L, Carrier L (May 2012). "How
do MYBPC3 mutations cause hypertrophic cardiomyopathy?". Journal of Muscle Research
and Cell Motility. 33 (1): 75–80. doi:10.1007/s10974-011-9268-3 (https://doi.org/10.1007%2
Fs10974-011-9268-3). PMID 22057632 (https://www.ncbi.nlm.nih.gov/pubmed/22057632).
80. van der Velden J, Ho CY, Tardiff JC, Olivotto I, Knollmann BC, Carrier L (Apr 2015).
"Research priorities in sarcomeric cardiomyopathies" (https://www.ncbi.nlm.nih.gov/pmc/arti
cles/PMC4375392). Cardiovascular Research. 105 (4): 449–56. doi:10.1093/cvr/cvv019 (htt
ps://doi.org/10.1093%2Fcvr%2Fcvv019). PMC 4375392 (https://www.ncbi.nlm.nih.gov/pmc/
articles/PMC4375392). PMID 25631582 (https://www.ncbi.nlm.nih.gov/pubmed/25631582).
81. Sarikas A, Carrier L, Schenke C, Doll D, Flavigny J, Lindenberg KS, Eschenhagen T, Zolk O
(Apr 2005). "Impairment of the ubiquitin-proteasome system by truncated cardiac myosin
binding protein C mutants". Cardiovascular Research. 66 (1): 33–44.
doi:10.1016/j.cardiores.2005.01.004 (https://doi.org/10.1016%2Fj.cardiores.2005.01.004).
PMID 15769446 (https://www.ncbi.nlm.nih.gov/pubmed/15769446).
82. Bahrudin U, Morisaki H, Morisaki T, Ninomiya H, Higaki K, Nanba E, Igawa O, Takashima S,
Mizuta E, Miake J, Yamamoto Y, Shirayoshi Y, Kitakaze M, Carrier L, Hisatome I (Dec
2008). "Ubiquitin-proteasome system impairment caused by a missense cardiac myosin-
binding protein C mutation and associated with cardiac dysfunction in hypertrophic
cardiomyopathy" (http://repository.lib.tottori-u.ac.jp/files/public/0/4136/20180622151113992
329/G14_15101A00024R.pdf) (PDF). Journal of Molecular Biology. 384 (4): 896–907.
doi:10.1016/j.jmb.2008.09.070 (https://doi.org/10.1016%2Fj.jmb.2008.09.070).
PMID 18929575 (https://www.ncbi.nlm.nih.gov/pubmed/18929575).
83. Vignier N, Schlossarek S, Fraysse B, Mearini G, Krämer E, Pointu H, Mougenot N, Guiard
J, Reimer R, Hohenberg H, Schwartz K, Vernet M, Eschenhagen T, Carrier L (Jul 2009).
"Nonsense-mediated mRNA decay and ubiquitin-proteasome system regulate cardiac
myosin-binding protein C mutant levels in cardiomyopathic mice". Circulation Research. 105
(3): 239–48. doi:10.1161/CIRCRESAHA.109.201251 (https://doi.org/10.1161%2FCIRCRES
AHA.109.201251). PMID 19590044 (https://www.ncbi.nlm.nih.gov/pubmed/19590044).
84. Mearini G, Schlossarek S, Willis MS, Carrier L (Dec 2008). "The ubiquitin-proteasome
system in cardiac dysfunction". Biochimica et Biophysica Acta. 1782 (12): 749–63.
doi:10.1016/j.bbadis.2008.06.009 (https://doi.org/10.1016%2Fj.bbadis.2008.06.009).
PMID 18634872 (https://www.ncbi.nlm.nih.gov/pubmed/18634872).
85. Carrier L, Schlossarek S, Willis MS, Eschenhagen T (Jan 2010). "The ubiquitin-proteasome
system and nonsense-mediated mRNA decay in hypertrophic cardiomyopathy" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC4023315). Cardiovascular Research. 85 (2): 330–8.
doi:10.1093/cvr/cvp247 (https://doi.org/10.1093%2Fcvr%2Fcvp247). PMC 4023315 (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC4023315). PMID 19617224 (https://www.ncbi.nlm.ni
h.gov/pubmed/19617224).
86. Schlossarek S, Frey N, Carrier L (Jun 2014). "Ubiquitin-proteasome system and hereditary
cardiomyopathies". Journal of Molecular and Cellular Cardiology. 71: 25–31.
doi:10.1016/j.yjmcc.2013.12.016 (https://doi.org/10.1016%2Fj.yjmcc.2013.12.016).
PMID 24380728 (https://www.ncbi.nlm.nih.gov/pubmed/24380728).
87. Gedicke-Hornung C, Behrens-Gawlik V, Reischmann S, Geertz B, Stimpel D, Weinberger F,
Schlossarek S, Précigout G, Braren I, Eschenhagen T, Mearini G, Lorain S, Voit T, Dreyfus
PA, Garcia L, Carrier L (Jul 2013). "Rescue of cardiomyopathy through U7snRNA-mediated
exon skipping in Mybpc3-targeted knock-in mice" (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC3721478). EMBO Molecular Medicine. 5 (7): 1128–1145.
doi:10.1002/emmm.201202168 (https://doi.org/10.1002%2Femmm.201202168).
PMC 3721478 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3721478). PMID 23716398
(https://www.ncbi.nlm.nih.gov/pubmed/23716398).
88. Mearini G, Stimpel D, Krämer E, Geertz B, Braren I, Gedicke-Hornung C, Précigout G,
Müller OJ, Katus HA, Eschenhagen T, Voit T, Garcia L, Lorain S, Carrier L (2013). "Repair of
Mybpc3 mRNA by 5'-trans-splicing in a Mouse Model of Hypertrophic Cardiomyopathy" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC3731888). Molecular Therapy: Nucleic Acids. 2:
e102. doi:10.1038/mtna.2013.31 (https://doi.org/10.1038%2Fmtna.2013.31). PMC 3731888
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3731888). PMID 23820890 (https://www.ncb
i.nlm.nih.gov/pubmed/23820890).
89. Vignier N, Schlossarek S, Fraysse B, Mearini G, Krämer E, Pointu H, Mougenot N, Guiard
J, Reimer R, Hohenberg H, Schwartz K, Vernet M, Eschenhagen T, Carrier L (Jul 2009).
"Nonsense-mediated mRNA decay and ubiquitin-proteasome system regulate cardiac
myosin-binding protein C mutant levels in cardiomyopathic mice". Circulation Research. 105
(3): 239–48. doi:10.1161/CIRCRESAHA.109.201251 (https://doi.org/10.1161%2FCIRCRES
AHA.109.201251). PMID 19590044 (https://www.ncbi.nlm.nih.gov/pubmed/19590044).
90. Schlossarek S, Englmann DR, Sultan KR, Sauer M, Eschenhagen T, Carrier L (Jan 2012).
"Defective proteolytic systems in Mybpc3-targeted mice with cardiac hypertrophy" (https://ze
nodo.org/record/3411902). Basic Research in Cardiology. 107 (1): 235.
doi:10.1007/s00395-011-0235-3 (https://doi.org/10.1007%2Fs00395-011-0235-3).
PMID 22189562 (https://www.ncbi.nlm.nih.gov/pubmed/22189562).
91. Schlossarek S, Schuermann F, Geertz B, Mearini G, Eschenhagen T, Carrier L (May 2012).
"Adrenergic stress reveals septal hypertrophy and proteasome impairment in heterozygous
Mybpc3-targeted knock-in mice" (https://zenodo.org/record/3413136). Journal of Muscle
Research and Cell Motility. 33 (1): 5–15. doi:10.1007/s10974-011-9273-6 (https://doi.org/10.
1007%2Fs10974-011-9273-6). PMID 22076249 (https://www.ncbi.nlm.nih.gov/pubmed/220
76249).
92. Predmore JM, Wang P, Davis F, Bartolone S, Westfall MV, Dyke DB, Pagani F, Powell SR,
Day SM (Mar 2010). "Ubiquitin proteasome dysfunction in human hypertrophic and dilated
cardiomyopathies" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2857348). Circulation.
121 (8): 997–1004. doi:10.1161/CIRCULATIONAHA.109.904557 (https://doi.org/10.1161%2
FCIRCULATIONAHA.109.904557). PMC 2857348 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC2857348). PMID 20159828 (https://www.ncbi.nlm.nih.gov/pubmed/20159828).
93. Witt CC, Gerull B, Davies MJ, Centner T, Linke WA, Thierfelder L (Feb 2001).
"Hypercontractile properties of cardiac muscle fibers in a knock-in mouse model of cardiac
myosin-binding protein-C". The Journal of Biological Chemistry. 276 (7): 5353–9.
doi:10.1074/jbc.M008691200 (https://doi.org/10.1074%2Fjbc.M008691200).
PMID 11096095 (https://www.ncbi.nlm.nih.gov/pubmed/11096095).
94. van Dijk SJ, Dooijes D, dos Remedios C, Michels M, Lamers JM, Winegrad S, Schlossarek
S, Carrier L, ten Cate FJ, Stienen GJ, van der Velden J (Mar 2009). "Cardiac myosin-
binding protein C mutations and hypertrophic cardiomyopathy: haploinsufficiency, deranged
phosphorylation, and cardiomyocyte dysfunction". Circulation. 119 (11): 1473–83.
doi:10.1161/CIRCULATIONAHA.108.838672 (https://doi.org/10.1161%2FCIRCULATIONAH
A.108.838672). PMID 19273718 (https://www.ncbi.nlm.nih.gov/pubmed/19273718).
95. Fraysse B, Weinberger F, Bardswell SC, Cuello F, Vignier N, Geertz B, Starbatty J, Krämer
E, Coirault C, Eschenhagen T, Kentish JC, Avkiran M, Carrier L (Jun 2012). "Increased
myofilament Ca2+ sensitivity and diastolic dysfunction as early consequences of Mybpc3
mutation in heterozygous knock-in mice" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC33
70652). Journal of Molecular and Cellular Cardiology. 52 (6): 1299–307.
doi:10.1016/j.yjmcc.2012.03.009 (https://doi.org/10.1016%2Fj.yjmcc.2012.03.009).
PMC 3370652 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3370652). PMID 22465693
(https://www.ncbi.nlm.nih.gov/pubmed/22465693).
96. van Dijk SJ, Paalberends ER, Najafi A, Michels M, Sadayappan S, Carrier L, Boontje NM,
Kuster DW, van Slegtenhorst M, Dooijes D, dos Remedios C, ten Cate FJ, Stienen GJ, van
der Velden J (Jan 2012). "Contractile dysfunction irrespective of the mutant protein in
human hypertrophic cardiomyopathy with normal systolic function". Circulation: Heart
Failure. 5 (1): 36–46. doi:10.1161/CIRCHEARTFAILURE.111.963702 (https://doi.org/10.116
1%2FCIRCHEARTFAILURE.111.963702). PMID 22178992 (https://www.ncbi.nlm.nih.gov/p
ubmed/22178992).
97. Sequeira V, Wijnker PJ, Nijenkamp LL, Kuster DW, Najafi A, Witjas-Paalberends ER, Regan
JA, Boontje N, Ten Cate FJ, Germans T, Carrier L, Sadayappan S, van Slegtenhorst MA,
Zaremba R, Foster DB, Murphy AM, Poggesi C, Dos Remedios C, Stienen GJ, Ho CY,
Michels M, van der Velden J (May 2013). "Perturbed length-dependent activation in human
hypertrophic cardiomyopathy with missense sarcomeric gene mutations" (https://www.ncbi.n
lm.nih.gov/pmc/articles/PMC3675884). Circulation Research. 112 (11): 1491–505.
doi:10.1161/CIRCRESAHA.111.300436 (https://doi.org/10.1161%2FCIRCRESAHA.111.300
436). PMC 3675884 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3675884).
PMID 23508784 (https://www.ncbi.nlm.nih.gov/pubmed/23508784).
98. Stöhr A, Friedrich FW, Flenner F, Geertz B, Eder A, Schaaf S, Hirt MN, Uebeler J,
Schlossarek S, Carrier L, Hansen A, Eschenhagen T (Oct 2013). "Contractile abnormalities
and altered drug response in engineered heart tissue from Mybpc3-targeted knock-in mice".
Journal of Molecular and Cellular Cardiology. 63: 189–98. doi:10.1016/j.yjmcc.2013.07.011
(https://doi.org/10.1016%2Fj.yjmcc.2013.07.011). PMID 23896226 (https://www.ncbi.nlm.ni
h.gov/pubmed/23896226).
99. Jung G, Bernstein D (Jul 2014). "hiPSC Modeling of Inherited Cardiomyopathies" (https://w
ww.ncbi.nlm.nih.gov/pmc/articles/PMC4096486). Current Treatment Options in
Cardiovascular Medicine. 16 (7): 320. doi:10.1007/s11936-014-0320-7 (https://doi.org/10.10
07%2Fs11936-014-0320-7). PMC 4096486 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC
4096486). PMID 24838688 (https://www.ncbi.nlm.nih.gov/pubmed/24838688).
100. Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, Han L, Yen M, Wang Y,
Sun N, Abilez OJ, Hu S, Ebert AD, Navarrete EG, Simmons CS, Wheeler M, Pruitt B, Lewis
R, Yamaguchi Y, Ashley EA, Bers DM, Robbins RC, Longaker MT, Wu JC (Jan 2013).
"Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy
pathology in patient-specific induced pluripotent stem cells" (https://www.ncbi.nlm.nih.gov/p
mc/articles/PMC3638033). Cell Stem Cell. 12 (1): 101–13. doi:10.1016/j.stem.2012.10.010
(https://doi.org/10.1016%2Fj.stem.2012.10.010). PMC 3638033 (https://www.ncbi.nlm.nih.g
ov/pmc/articles/PMC3638033). PMID 23290139 (https://www.ncbi.nlm.nih.gov/pubmed/232
90139).
101. Han L, Li Y, Tchao J, Kaplan AD, Lin B, Li Y, Mich-Basso J, Lis A, Hassan N, London B, Bett
GC, Tobita K, Rasmusson RL, Yang L (Nov 2014). "Study familial hypertrophic
cardiomyopathy using patient-specific induced pluripotent stem cells" (https://www.ncbi.nlm.
nih.gov/pmc/articles/PMC4217687). Cardiovascular Research. 104 (2): 258–69.
doi:10.1093/cvr/cvu205 (https://doi.org/10.1093%2Fcvr%2Fcvu205). PMC 4217687 (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC4217687). PMID 25209314 (https://www.ncbi.nlm.ni
h.gov/pubmed/25209314).
102. Tanaka A, Yuasa S, Mearini G, Egashira T, Seki T, Kodaira M, Kusumoto D, Kuroda Y,
Okata S, Suzuki T, Inohara T, Arimura T, Makino S, Kimura K, Kimura A, Furukawa T,
Carrier L, Node K, Fukuda K (Dec 2014). "Endothelin-1 induces myofibrillar disarray and
contractile vector variability in hypertrophic cardiomyopathy-induced pluripotent stem cell-
derived cardiomyocytes" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4338713). Journal
of the American Heart Association. 3 (6): e001263. doi:10.1161/JAHA.114.001263 (https://d
oi.org/10.1161%2FJAHA.114.001263). PMC 4338713 (https://www.ncbi.nlm.nih.gov/pmc/art
icles/PMC4338713). PMID 25389285 (https://www.ncbi.nlm.nih.gov/pubmed/25389285).
103. Tanaka A, Yuasa S, Mearini G, Egashira T, Seki T, Kodaira M, Kusumoto D, Kuroda Y,
Okata S, Suzuki T, Inohara T, Arimura T, Makino S, Kimura K, Kimura A, Furukawa T,
Carrier L, Node K, Fukuda K (Dec 2014). "Endothelin-1 induces myofibrillar disarray and
contractile vector variability in hypertrophic cardiomyopathy-induced pluripotent stem cell-
derived cardiomyocytes" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4338713). Journal
of the American Heart Association. 3 (6): e001263. doi:10.1161/JAHA.114.001263 (https://d
oi.org/10.1161%2FJAHA.114.001263). PMC 4338713 (https://www.ncbi.nlm.nih.gov/pmc/art
icles/PMC4338713). PMID 25389285 (https://www.ncbi.nlm.nih.gov/pubmed/25389285).
104. Zacchigna S, Zentilin L, Giacca M (May 2014). "Adeno-associated virus vectors as
therapeutic and investigational tools in the cardiovascular system". Circulation Research.
114 (11): 1827–46. doi:10.1161/CIRCRESAHA.114.302331 (https://doi.org/10.1161%2FCIR
CRESAHA.114.302331). PMID 24855205
(https://www.ncbi.nlm.nih.gov/pubmed/24855205).
105. Hammond SM, Wood MJ (May 2011). "Genetic therapies for RNA mis-splicing diseases".
Trends in Genetics. 27 (5): 196–205. doi:10.1016/j.tig.2011.02.004 (https://doi.org/10.101
6%2Fj.tig.2011.02.004). PMID 21497936 (https://www.ncbi.nlm.nih.gov/pubmed/21497936).
106. Doudna JA, Charpentier E (Nov 2014). "Genome editing. The new frontier of genome
engineering with CRISPR-Cas9". Science. 346 (6213): 1258096.
doi:10.1126/science.1258096 (https://doi.org/10.1126%2Fscience.1258096).
PMID 25430774 (https://www.ncbi.nlm.nih.gov/pubmed/25430774).
107. Hsu PD, Lander ES, Zhang F (Jun 2014). "Development and applications of CRISPR-Cas9
for genome engineering" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4343198). Cell.
157 (6): 1262–78. doi:10.1016/j.cell.2014.05.010 (https://doi.org/10.1016%2Fj.cell.2014.05.
010). PMC 4343198 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4343198).
PMID 24906146 (https://www.ncbi.nlm.nih.gov/pubmed/24906146).
108. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F (Nov 2013). "Genome
engineering using the CRISPR-Cas9 system" (https://www.ncbi.nlm.nih.gov/pmc/articles/PM
C3969860). Nature Protocols. 8 (11): 2281–308. doi:10.1038/nprot.2013.143 (https://doi.or
g/10.1038%2Fnprot.2013.143). PMC 3969860 (https://www.ncbi.nlm.nih.gov/pmc/articles/P
MC3969860). PMID 24157548 (https://www.ncbi.nlm.nih.gov/pubmed/24157548).
109. Woodley L, Valcárcel J (Oct 2002). "Regulation of alternative pre-mRNA splicing". Briefings
in Functional Genomics & Proteomics. 1 (3): 266–77. doi:10.1093/bfgp/1.3.266 (https://doi.o
rg/10.1093%2Fbfgp%2F1.3.266). PMID 15239893 (https://www.ncbi.nlm.nih.gov/pubmed/1
5239893).
110. Goyenvalle A, Vulin A, Fougerousse F, Leturcq F, Kaplan JC, Garcia L, Danos O (Dec
2004). "Rescue of dystrophic muscle through U7 snRNA-mediated exon skipping". Science.
306 (5702): 1796–9. doi:10.1126/science.1104297 (https://doi.org/10.1126%2Fscience.110
4297). PMID 15528407 (https://www.ncbi.nlm.nih.gov/pubmed/15528407).
111. Wally V, Murauer EM, Bauer JW (Aug 2012). "Spliceosome-mediated trans-splicing: the
therapeutic cut and paste". The Journal of Investigative Dermatology. 132 (8): 1959–66.
doi:10.1038/jid.2012.101 (https://doi.org/10.1038%2Fjid.2012.101). PMID 22495179 (http
s://www.ncbi.nlm.nih.gov/pubmed/22495179).
112. Mearini G, Stimpel D, Geertz B, Weinberger F, Krämer E, Schlossarek S, Mourot-Filiatre J,
Stoehr A, Dutsch A, Wijnker PJ, Braren I, Katus HA, Müller OJ, Voit T, Eschenhagen T,
Carrier L (2014). "Mybpc3 gene therapy for neonatal cardiomyopathy enables long-term
disease prevention in mice". Nature Communications. 5: 5515. doi:10.1038/ncomms6515 (h
ttps://doi.org/10.1038%2Fncomms6515). PMID 25463264 (https://www.ncbi.nlm.nih.gov/pu
bmed/25463264).
113. Duncker DJ, Bakkers J, Brundel BJ, Robbins J, Tardiff JC, Carrier L (Apr 2015). "Animal
and in silico models for the study of sarcomeric cardiomyopathies" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC4375391). Cardiovascular Research. 105 (4): 439–48.
doi:10.1093/cvr/cvv006 (https://doi.org/10.1093%2Fcvr%2Fcvv006). PMC 4375391 (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC4375391). PMID 25600962 (https://www.ncbi.nlm.ni
h.gov/pubmed/25600962).

Further reading
Vikstrom KL, Leinwand LA (Feb 1996). "Contractile protein mutations and heart disease".
Current Opinion in Cell Biology. 8 (1): 97–105. doi:10.1016/S0955-0674(96)80053-6 (http
s://doi.org/10.1016%2FS0955-0674%2896%2980053-6). PMID 8791411 (https://www.ncbi.
nlm.nih.gov/pubmed/8791411).
Schaub MC, Hefti MA, Zuellig RA, Morano I (Feb 1998). "Modulation of contractility in
human cardiac hypertrophy by myosin essential light chain isoforms" (http://doc.rero.ch/reco
rd/301870/files/37-2-381.pdf) (PDF). Cardiovascular Research. 37 (2): 381–404.
doi:10.1016/S0008-6363(97)00258-7 (https://doi.org/10.1016%2FS0008-6363%2897%2900
258-7). PMID 9614495 (https://www.ncbi.nlm.nih.gov/pubmed/9614495).
Bonne G, Carrier L, Richard P, Hainque B, Schwartz K (Sep 1998). "Familial hypertrophic
cardiomyopathy: from mutations to functional defects". Circulation Research. 83 (6): 580–
93. doi:10.1161/01.res.83.6.580 (https://doi.org/10.1161%2F01.res.83.6.580).
PMID 9742053 (https://www.ncbi.nlm.nih.gov/pubmed/9742053).
Jääskeläinen P, Miettinen R, Kärkkäinen P, Toivonen L, Laakso M, Kuusisto J (2004).
"Genetics of hypertrophic cardiomyopathy in eastern Finland: few founder mutations with
benign or intermediary phenotypes". Annals of Medicine. 36 (1): 23–32.
doi:10.1080/07853890310017161 (https://doi.org/10.1080%2F07853890310017161).
PMID 15000344 (https://www.ncbi.nlm.nih.gov/pubmed/15000344).
Starr R, Offer G (Jun 1978). "The interaction of C-protein with heavy meromyosin and
subfragment-2" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1184031). The Biochemical
Journal. 171 (3): 813–6. doi:10.1042/bj1710813 (https://doi.org/10.1042%2Fbj1710813).
PMC 1184031 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1184031). PMID 352343 (htt
ps://www.ncbi.nlm.nih.gov/pubmed/352343).
Moos C, Feng IN (Oct 1980). "Effect of C-protein on actomyosin ATPase". Biochimica et
Biophysica Acta. 632 (2): 141–9. doi:10.1016/0304-4165(80)90071-9 (https://doi.org/10.101
6%2F0304-4165%2880%2990071-9). PMID 6448079 (https://www.ncbi.nlm.nih.gov/pubme
d/6448079).
Watkins H, Conner D, Thierfelder L, Jarcho JA, MacRae C, McKenna WJ, Maron BJ,
Seidman JG, Seidman CE (Dec 1995). "Mutations in the cardiac myosin binding protein-C
gene on chromosome 11 cause familial hypertrophic cardiomyopathy". Nature Genetics. 11
(4): 434–7. doi:10.1038/ng1295-434 (https://doi.org/10.1038%2Fng1295-434).
PMID 7493025 (https://www.ncbi.nlm.nih.gov/pubmed/7493025).
Bonne G, Carrier L, Bercovici J, Cruaud C, Richard P, Hainque B, Gautel M, Labeit S,
James M, Beckmann J, Weissenbach J, Vosberg HP, Fiszman M, Komajda M, Schwartz K
(Dec 1995). "Cardiac myosin binding protein-C gene splice acceptor site mutation is
associated with familial hypertrophic cardiomyopathy". Nature Genetics. 11 (4): 438–40.
doi:10.1038/ng1295-438 (https://doi.org/10.1038%2Fng1295-438). PMID 7493026 (https://w
ww.ncbi.nlm.nih.gov/pubmed/7493026).
Gautel M, Zuffardi O, Freiburg A, Labeit S (May 1995). "Phosphorylation switches specific
for the cardiac isoform of myosin binding protein-C: a modulator of cardiac contraction?" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294). The EMBO Journal. 14 (9): 1952–60.
doi:10.1002/j.1460-2075.1995.tb07187.x (https://doi.org/10.1002%2Fj.1460-2075.1995.tb07
187.x). PMC 398294 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC398294).
PMID 7744002 (https://www.ncbi.nlm.nih.gov/pubmed/7744002).
Carrier L, Hengstenberg C, Beckmann JS, Guicheney P, Dufour C, Bercovici J, Dausse E,
Berebbi-Bertrand I, Wisnewsky C, Pulvenis D (Jul 1993). "Mapping of a novel gene for
familial hypertrophic cardiomyopathy to chromosome 11". Nature Genetics. 4 (3): 311–3.
doi:10.1038/ng0793-311 (https://doi.org/10.1038%2Fng0793-311). PMID 8358441 (https://w
ww.ncbi.nlm.nih.gov/pubmed/8358441).
Freiburg A, Gautel M (Jan 1996). "A molecular map of the interactions between titin and
myosin-binding protein C. Implications for sarcomeric assembly in familial hypertrophic
cardiomyopathy". European Journal of Biochemistry / FEBS. 235 (1–2): 317–23.
doi:10.1111/j.1432-1033.1996.00317.x (https://doi.org/10.1111%2Fj.1432-1033.1996.0031
7.x). PMID 8631348 (https://www.ncbi.nlm.nih.gov/pubmed/8631348).
Carrier L, Bonne G, Bährend E, Yu B, Richard P, Niel F, Hainque B, Cruaud C, Gary F,
Labeit S, Bouhour JB, Dubourg O, Desnos M, Hagège AA, Trent RJ, Komajda M, Fiszman
M, Schwartz K (Mar 1997). "Organization and sequence of human cardiac myosin binding
protein C gene (MYBPC3) and identification of mutations predicted to produce truncated
proteins in familial hypertrophic cardiomyopathy". Circulation Research. 80 (3): 427–34.
doi:10.1161/01.res.0000435859.24609.b3 (https://doi.org/10.1161%2F01.res.0000435859.2
4609.b3). PMID 9048664 (https://www.ncbi.nlm.nih.gov/pubmed/9048664).
Rottbauer W, Gautel M, Zehelein J, Labeit S, Franz WM, Fischer C, Vollrath B, Mall G, Dietz
R, Kübler W, Katus HA (Jul 1997). "Novel splice donor site mutation in the cardiac myosin-
binding protein-C gene in familial hypertrophic cardiomyopathy. Characterization Of cardiac
transcript and protein" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC508212). The Journal
of Clinical Investigation. 100 (2): 475–82. doi:10.1172/JCI119555 (https://doi.org/10.1172%
2FJCI119555). PMC 508212 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC508212).
PMID 9218526 (https://www.ncbi.nlm.nih.gov/pubmed/9218526).
Yu B, French JA, Carrier L, Jeremy RW, McTaggart DR, Nicholson MR, Hambly B,
Semsarian C, Richmond DR, Schwartz K, Trent RJ (Mar 1998). "Molecular pathology of
familial hypertrophic cardiomyopathy caused by mutations in the cardiac myosin binding
protein C gene" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1051243). Journal of
Medical Genetics. 35 (3): 205–10. doi:10.1136/jmg.35.3.205 (https://doi.org/10.1136%2Fjm
g.35.3.205). PMC 1051243 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1051243).
PMID 9541104 (https://www.ncbi.nlm.nih.gov/pubmed/9541104).
Moolman-Smook JC, Mayosi B, Brink P, Corfield VA (Mar 1998). "Identification of a new
missense mutation in MyBP-C associated with hypertrophic cardiomyopathy" (https://www.n
cbi.nlm.nih.gov/pmc/articles/PMC1051254). Journal of Medical Genetics. 35 (3): 253–4.
doi:10.1136/jmg.35.3.253 (https://doi.org/10.1136%2Fjmg.35.3.253). PMC 1051254 (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC1051254). PMID 9541115 (https://www.ncbi.nlm.ni
h.gov/pubmed/9541115).
Niimura H, Bachinski LL, Sangwatanaroj S, Watkins H, Chudley AE, McKenna W,
Kristinsson A, Roberts R, Sole M, Maron BJ, Seidman JG, Seidman CE (Apr 1998).
"Mutations in the gene for cardiac myosin-binding protein C and late-onset familial
hypertrophic cardiomyopathy". The New England Journal of Medicine. 338 (18): 1248–57.
doi:10.1056/NEJM199804303381802
(https://doi.org/10.1056%2FNEJM199804303381802). PMID 9562578 (https://www.ncbi.nl
m.nih.gov/pubmed/9562578).
Richard P, Isnard R, Carrier L, Dubourg O, Donatien Y, Mathieu B, Bonne G, Gary F,
Charron P, Hagege M, Komajda M, Schwartz K, Hainque B (Jul 1999). "Double
heterozygosity for mutations in the beta-myosin heavy chain and in the cardiac myosin
binding protein C genes in a family with hypertrophic cardiomyopathy" (https://www.ncbi.nl
m.nih.gov/pmc/articles/PMC1734410). Journal of Medical Genetics. 36 (7): 542–5.
doi:10.1136/jmg.36.7.542 (https://doi.org/10.1136%2Fjmg.36.7.542) (inactive 2019-12-03).
PMC 1734410 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1734410). PMID 10424815
(https://www.ncbi.nlm.nih.gov/pubmed/10424815).

External links
Mass spectrometry characterization of MYBPC3 at COPaKB (http://www.heartproteome.or
g/copa/ProteinInfo.aspx?QType=Protein%20ID&QValue=Q14896)
GeneReviews/NIH/NCBI/UW entry on Familial Hypertrophic Cardiomyopathy Overview (http
s://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=gene&part=hyper-card)
Overview of all the structural information available in the PDB for UniProt: Q14896 (https://w
ww.ebi.ac.uk/pdbe/pdbe-kb/proteins/Q14896) (Myosin-binding protein C, cardiac-type) at
the PDBe-KB.
Retrieved from "https://en.wikipedia.org/w/index.php?title=Myosin_binding_protein_C,_cardiac&oldid=929096310"

This page was last edited on 3 December 2019, at 17:07 (UTC).

Text is available under the Creative Commons Attribution-ShareAlike License; additional terms may apply. By using
this site, you agree to the Terms of Use and Privacy Policy. Wikipedia® is a registered trademark of the Wikimedia
Foundation, Inc., a non-profit organization.

You might also like