ArticlePDF Available

Abstract

Mutations in Lamin A/C gene (lmna) cause a wide spectrum of cardiolaminopathies strictly associated with significant deterioration of the electrical and contractile function of the heart. Despite the continuous flow of biomedical evidence, linking cardiac inflammation to heart remodelling in patients harbouring lmna mutations is puzzling. Therefore, we profiled 30 serum cytokines/chemokines in patients belonging to four different families carrying pathogenic lmna mutations segregating with cardiac phenotypes at different stages of severity (n = 19) and in healthy subjects (n = 11). Regardless lmna mutation subtype, high levels of circulating granulocyte colony‐stimulating factor (G‐CSF) and interleukin 6 (IL‐6) were found in all affected patients’ sera. In addition, elevated levels of Interleukins (IL) IL‐1Ra, IL‐1β IL‐4, IL‐5 and IL‐8 and the granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) were measured in a large subset of patients associated with more aggressive clinical manifestations. Finally, the expression of the pro‐inflammatory 70 kDa heat shock protein (Hsp70) was significantly increased in serum exosomes of patients harbouring the lmna mutation associated with the more severe phenotype. Overall, the identification of patient subsets with overactive or dysregulated myocardial inflammatory responses could represent an innovative diagnostic, prognostic and therapeutic tool against Lamin A/C cardiomyopathies.
10902
|
J Cell Mol Med. 2021;25:10902–10915.wileyonlinelibrary.com/journal/jcmm
1 | INTRODUCTION
Mutations in lmna gene encoding intermediate filament proteins of
the inner n uclear membra ne Lamin A/C (LMNA ) cause tissue- specif ic
systemic diseases collectively known as laminopathies. Cardiac lami-
nopathies encompass a wide spectrum of clinical entities with high
penetrance and (usually) adult onset. Although dilated cardiomyopa-
thy with conduction defects (DCM- CD) is the most prevalent pheno-
type, different mutational sites might correlate with different clinical
manifestations spanning from conduction disorders, frequent atrial
fibrillation and life- threatening ventricular arrhythmias, with nor-
mal or altered ventricular systolic function.1,2 This surprising large
Received: 22 July 20 21 
|
Revised: 16 Septemb er 2021 
|
Accepted: 23 September 2021
DOI: 10.1111/jcmm.16975
ORIGINAL ARTICLE
Pro- inflammatory cytokines as emerging molecular
determinants in cardiolaminopathies
Andrea Gerbino1| Cinzia Forleo2| Serena Milano1| Francesca Piccapane1|
Giuseppe Procino1| Martino Pepe2| Mara Piccolo2| Piero Guida3| Nicoletta Resta4|
Stefano Favale2| Maria Svelto1| Monica Carmosino5
This is an op en acces s article unde r the terms of the Creat ive Commo ns At tri bution License, which permits use, distr ibution and reproduc tion in any medium ,
provide d the original wor k is properly cited.
© 2021 The Authors. Journal of Cellular and Molecular Medicine publish ed by Foundation fo r Cellular and Molecular Medicine and Joh n Wiley & Sons Ltd.
Gerbi no and Forleo con tribut ed equally to thi s work as first au thors.
1Department of Biosciences,
Biotechnologies and Biopharmaceutics,
University of Ba ri, Bar i, Ital y
2Department of Emergency and Organ
Transplantation, Cardiology Unit,
University of Ba ri Aldo M oro, Bar i, Ital y
3Regional Gener al Hospital “F. Miulli”,
Acquaviva delle Fonti, Italy
4Division of Medical Genetics,
Depar tment of B iomedical Sciences and
Human On colog y, University of Bar i Aldo
Moro, Bari, Italy
5Depar tment of S cience s, Unive rsity of
Basilicata, Potenza, Italy
Correspondence
Monica Carmosino, Department of
Science s, Unive rsity of Basilicata, Potenza,
Italy.
Email: monica.carmosino@unibas.it
Funding information
This work w as supported by funding
from ‘Carmosino19LAMINOPATIE’ and
‘Carmosino20RIL’ to Monica Carmosino
and from t he CLUSTER TECNOLOG ICO
REGIONALE ‘DICLIMA X’ (project #
MTJU9H8) to Maria Svelto
Abstract
Mutations in Lamin A/C gene (lmna) cause a wide spectrum of cardiolaminopathies
strictly associated with significant deterioration of the electrical and contractile
function of the heart. Despite the continuous flow of biomedical evidence, linking
cardiac inflammation to heart remodelling in patients harbouring lmna mutations is
puzzling. Therefore, we profiled 30 serum cytokines/chemokines in patients belong-
ing to four different families carr ying pathogenic lmna mutations segregating with
cardiac phenotypes at different stages of severity (n = 19) and in healthy subjects (n =
11). Regardless lmna mutation subtype, high levels of circulating granulocyte colony-
stimulating factor (G- CSF) and interleukin 6 (IL- 6) were found in all affected patients’
sera. In addition, elevated levels of Interleukins (IL) IL- 1Ra, IL- 1β IL- 4, IL- 5 and IL- 8
and the granulocyte- macrophage colony- stimulating factor (GM- CSF) were measured
in a large subset of patients associated with more aggressive clinical manifestations.
Finally, the expression of the pro- inflammatory 70 kDa heat shock protein (Hsp70)
was significantly increased in serum exosomes of patients harbouring the lmna muta-
tion associated with the more severe phenotype. Overall, the identification of patient
subsets with overactive or dysregulated myocardial inflammator y responses could
represent an innovative diagnostic, prognostic and therapeutic tool against Lamin
A/C cardiomyopathies.
KEYWORDS
cardiolaminopathies, cytokines, inflammation
   
|
10903
GERBIN O Et al.
number of cardiac phenotypes reflects an even larger number of
specific mutations (about 500) identified in the lmna gene and col-
lected in the UMD- lmna mutations database.
The molecular mechanisms underlying the origin and develop-
ment of such a wide spectrum of cardiac pathologies are still in-
completely defined. Despite under the spotlight of many research
groups for quite a long time, a complete understanding of the cellular
functions mediated by LMNA is missing. Thus, mutations in the lmna
gene can either affect a specific function of LMNA or broadly impact
the whole repertoire of molecular mechanisms controlled by these
intermediate filaments such as mechanotransduction, gene regula-
tion and signal transduction.3,4 Yet, we still do not know whether
these different pathogenic mechanisms represent different pieces
of the same puzzle or each one of them can be correlated to a spe-
cific clinical entity.
For instance, alterations in several cellular pathways, including
WNT/β- catenin, mitogen- activated protein kinases (MAPKs) and pro-
tein kinas e B (AKT)/mammal ian target of rapamy cin (mTOR) signall ing,
have been identified in LMNAH222P/H222P mice, a mouse model of the
human LMNA- associated DCM- CD.5,6 Interestingly, lmna mutations
may also impinge the machinery involved in C a2+ handling into the
ER7 9 and the connexin 43 (CX43) expression/activity at the plasma
membrane in cardiomyopathies.10,11 More recently, Salvarini et al.
demonstrated that the epigenetic inhibition of the sodium voltage-
gated channel alpha subunit 5 (SCN5A) in cardiomyocytes differen-
tiated from IPS derived from patients with K219T- LMNA pathogenic
variant12 can account for the conduction defects reported in the clin-
ical history of these arrhythmogenic patients.
Despite the molecular mechanism involved in LMNA cardiomy-
opathies, the deterioration in electrical and contractile function cor-
relates with exacerbated cardiomyocytes damage or death, which,
in turn, may trigger myocardial inflammation, further aggravating
the progression of the cardiomyopathy. Myocardial fibrosis is also
thought to be responsible for the development of both electrical in-
stability and mechanical impairment in cardiac laminopathies13 typi-
cally developing within the interventricular septum, near the region of
the conduction system thus accounting for the conduction disease.14
It is known that the mammalian heart contains a population of resi-
dent macrophages that proliferate following myocardial damage such
as in myocardial infarction, which in turn recruit other monocytes to
the hear t, contributing to myocardial interstitial fibrosis and adverse
cardiac remodelling.15 Moreover, it also well established that pro-
inflammatory cytokines are produced in typical inflammatory cardio-
myopathies as consequence of pathogen infection and a wide variety
of toxic substances, drugs and systemic immune- mediated diseases.
These cardiomyopathies evolve also in DCM and heart failure.16
However, in inherited cardiomyopathies, cardiac inflammation
and its correlation to contractile dysfunc tion and cardiac remodel-
ling have not been fully investigated. Therefore, in this study, we aim
at investigating the specific profile of 30 cytokines and chemokines
in the serum of four dif ferent groups of patients harbouring differ-
ent pathogenic mutations in lmna gene and their family members
not carr ying the mutation. The clinical phenotype associated with
the lmna mutations under investigation was mainly characterized by
left ventricular dilation, left ventricular systolic dysfunction and con-
duction defects despite at different range of severity, even amongst
members of the same family. The profile of inflammator y cytokines
measured in our experiments did not show any specificity for the
location of each mut ation analysed. However, the severity of the
clinical manifestations associated with each patient correlates with
the degree of inflammation in terms of numbers of pro- inflammatory
cytokines upregulated. Since no specific pharmacological treatment
is available and the ICD implantation is currently the only ef fective
clinical approach for patients affected by cardiac laminopathies,
the identification of patient subsets affected by LMNA cardiomy-
opathies with overactive or dysregulated myocardial inflammatory
responses could be crucial for clarifying the pathogenesis of these
cardiomyopathies and for evaluating immediate successful thera-
peutic approaches.
2 | METHODS
2.1  | Clinical and instrumental analysis
The patients who were referred to the Cardiomyopathy
Unit, Cardiolog y Unit, Department of Emergency and Organ
Transplantation, University of Bari Aldo Moro, Bari (Italy), between
January 2020 and August 2020, and who fulfilled the inclusion and
exclusion criteria, were involved in the study. A total of 30 Italian
patients (19 patients with lmna mutation e and 11 lmna mutation-
negative family members) were enrolled in the study and subjected
to blood sampling for serum cytokine assay. To avoid variations in
serum levels of cytokines, no subject had exercised physical activity
prior to blood sampling and did not have any ongoing infections or
immunodeficiency conditions at the time of enrolment. For further
details on inclusion/exclusion criteria, see Appendix S1. All recruited
subjects provided their written informed consent to participate in
this stu dy. The proj ect conformed to th e principles of the D eclaration
of Helsinki (World Medical Association) and was approved by the
Ethics Committee of the University Hospital Consortium, Policlinico
of Bari, Italy.
2.2  | Cytokine/chemokine assay
Plasma samples from the patients included in the study were pre-
pared by centrifugation at 500 g for 12 min and stored at −80 °C.
Bio- Plex Pro Human Cytokine 27- plex Assay (#M500KCAF0Y; Bio-
Rad Laboratories) and Bio- Plex Pro Transforming Growth Factor- β
(TGF- β) 3- plex Assay (#171W4001 M; Bio- Rad Laboratories) were
used by following the manufacturer's instructions. Each sample was
analysed in triplicate in BioPlexMagpix Multiplex Reader (Bio- Rad
Laboratories), and the data automatically analysed using Bio- Plex
Manager 6.0 software (Bio- Rad Laboratories). For the list of cy-
tokines analysed and for detailed procedure, see Appendix S1.
10904 
|
    G ERBINO E t al.
2.3  | Serum exosome preparation and analysis
Serum exosomes were isolated from 250 μl of each patient s’ serum
with the exosome isolation kit EXOQ50A- 1 (System Biosciences) ac-
cording to manufacturer's instructions. Serum exosomes were ana-
lysed by Western blotting for the expression of Hsp70 and exosome
markers. For details on serum exosomes preparation and analysis,
see Appendix S1.
2.4  | Cell culture and western blotting
HEK293 cells were transiently transfected with the previously de-
scribed17 vectors for the expression of either LMNA WT or LMNA- p.
Leu140_Ala146dup variant, using Lipofec tamine® 2000. After 72 h,
transfection cells were lysed in RIPA buf fer and lysate were analysed
by Western blotting for the expression of Hsp70. For details, see
Appendix S1.
2.5  | Statistical analysis
Continuous variables are expressed as mean values ± standard
deviation and compared between groups by using Student 's t test
(equal or unequal variance as appropriate). Categorical variables are
expressed as absolute frequency or percentage. Associations were
tested with Fisher's exact test. Analyses were performed using
STATA sof tware version 14 (Stata). Student's t test for unpaired data
was used to analyse dif ferences in cytokines levels bet ween each
patient group and controls. Receiver operating characteristic (ROC)
curves were used to estimate the diagnostic potential of the quanti-
fied individual cytokines to discriminate between groups. GraphPad
Prism software (version 8) was used for st atistical and graphical art s.
In all cases, significance was considered at p < 0.05.
3 | RESULTS
3.1  | Molecular analysis of the patients’ population
The lmna mutations included in this study were identified in mem-
bers of Italian families with cardiac phenotypes screened in our
Clinical Unit dedicated to cardiomyopathies.
The lmna mutations are as follows:
1. The heteroz ygous variant c.418_438dup consists of a duplication
of 21 nucleotides (CTGC TGAACTCCA AGGAGGCC) in the exon
2 of the lmna gene, located in the coil 1B of the central α-
helical rod domain of the LMNA protein. The resulting LMNA
variant is predicted to result in the duplication of seven amino
acids (LLNSKEA) in LMNA protein, from Leucine at position
140 to Alanine at position 146, without a frame shift in the
open reading frame. This LMNA variant hereinafter referred
to as p.Leu140_Ala146dup has been previously identified and
characterized in vitro by our group17 and classified as pathogenic.
2. The heterozygous variant c.329delG consists of a G deletion at
position 329 in the exon 2 of the lmna gene. This deletion causes
a shift in the reading frame starting at Arginine 110, changing
it to a Leucine and creating a premature stop codon at position
7 of the new reading frame. This LMNA variant, denoted as
p.Arg110Leufs*7, is present in the ClinVar data base and ranked
as likely pathogenic.
3. The heterozygous variant c.949G>A consists of a G to A substitu-
tion at position 949 in the exon 6 of the lmna gene leading to glu-
tamate to lysine exchange at the position 317 in the coil2 domain.
This variant (p.Glu317Lys) is ranked as pathogenic and has been
already described in patients with atrioventricular block (AVB)
and DCM.18,19
4. The heterozygous variant c.569G>A, located in coding exon 3 of
the lmna gene, results from a G to A substitution at nucleotide po-
sition 569. The arginine at codon 190 is replaced by glutamine, an
amino acid with highly similar properties. This variant is reported
as p.Arg190Gln and ranked as pathogenic. This alteration has
been already reported in individuals with DCM and reported as
LMNA R190Q variant.20,21
3.2  | Clinical data
Table 1 summarizes the most relevant clinical manifestations of all
the subjects evaluated in this study. Carriers of pathogenic LMNA
variants belonging to four different families (patient s) were com-
pared with family members not carr ying the mutation (controls).
Clinical data, electrical and mechanical abnormalities documented
by ECG recordings and cardiac imaging manifested during their clini-
cal history were reported.
3.2.1  |  Family 1
Patients harbouring p. Leu140_Ala146dup LMNA variant mostly
exhibited (4 out of 6) a phenotype charac terized by left ventricular
dilation (LVEDD 52 ± 6 mm; LVEDVi 74 ± 16 ml/m2), left ventricular
systolic dysfunction (LVEF 45 ± 13%) (Figure 1AD) and numerous
arrhythmic disorders. Indeed, frequent (up to 1000 per day) polymor-
phic premature ventricular complexes (PVCs), and non- sustained ven-
tricular tachycardia (NSVT) episodes were recorded on ECG Holter
monitoring in all patients of this group (Figure 2A); three patients also
showed sustained ventricular tachycardia (SVT) or ventricular fibrilla-
tion (VF) events, detected on telemetry or implantable cardioverter-
defibrillator (ICD) arrhythmia registry. Conduction disturbances (first- ,
second- and third- degree atrioventricular blocks, Figure 2B) and atrial
fibrillation (Figure 2C) were obser ved in about 80% of the patients.
Five patients received an ICD in primary prevention, and several de-
vice appropriate interventions by antit achycardia pacing (ATP) or DC
shock on SVT or fast SVT recognized in VF zone, respectively, were
   
|
10905
GERBIN O Et al.
TABLE 1 Clinical characteristics of study subjects according to lmna mutation carrier status
lmna mutation- positive subjects according to different genotypes (n = 19)
All lmna mutation-
positive subjects
(n = 19)
lmna mutation-
negative subjects
(n = 11)
p valuep. Leu140_Ala146dup (n = 6) p. Arg110Leuf s*7 (n = 4) p. Glu317Lys (n = 6) p. Arg190Gln (n = 3) Patients (n = 19) Controls (n = 11)
Age at time of blood
collection (years)
46 ± 10 42 ± 13 54 ± 6 35 ± 16 46 ± 12 41 ± 18 0.399
Gender (F/M) 4/2 3/1 2/4 0/3 9/10 7/4 0.466
Cardiac phenotype DCM, left ventricular systolic
dysfunction, AF, AVB,
PVCs, NSVT, SVT
Left ventricular systolic
dysfunction, AF, AVB,
PVCs, NSVT
DCM, lef t ventricular
systolic dysfunction, AF,
AVB, PVCs, NS VT, SVT
DCM, Lef t ventricle
systolic
dysfunction, AF
Healthy
Clinical myopathy 0 0 0 3 3 0
CPK levels (U/L) 163 ± 53 112 ± 68 59 ± 31 1274 ± 1255 339 ± 655 NA
SBP (mm Hg) 104 ± 13 106 ± 11 126 ± 20 153 ± 24 119 ± 24 118 ± 9 0.822
DBP (mm Hg) 65 ± 5 66 ± 8 83 ± 9 90 ± 13 75 ± 13 71 ± 7 0.320
BMI (K g/m2)24.9 ± 3.3 27. 1 ± 6.9 23.7 ± 3.4 27. 5 ± 4.3 25.4 ± 4.3 24. 3 ± 2.3 0.427
Heart rate (bpm) 60 ± 7 61 ± 11 62 ± 6 57 ± 2 60 ±773 ± 13 0.010
PR interval (ms) 250 ± 62 210 ± 68 277 ± 178 167± 23 237 ± 111 147± 29 0.003
QRS duration (ms) 93 ± 8 89 ± 10 97 ± 10 107 ± 12 96 ±11 87 ± 10 0.026
QTc interval (ms) 417 ± 24 398 ± 17 421 ± 22 410 ± 47 413 ± 26 415 ± 16 0.816
AF, n (%) 5 (83%) 2 (50%) 3 (50%) 1 (33%) 11 (58%) 0 (0%) 0.0 02
AV block, n (%) 5 (83%) 2 (50%) 3 (50%) 0 (0%) 10 (53%) 0 (0%) 0.004
PVCs >50 0/24h , n (%) 6 (100%) 3 (75%) 2 (33.3%) 0 (0%) 11 (58%) 0 (0%) 0.002
PVCs >1000/24h, n (%) 6 (100%) 3 (75%) 1 (16.6%) 0 (0%) 10 (53%) 0 (0%) 0.004
NS V T, n (%) 6 (100%) 2 (50%) 3 (50%) 0 (0%) 11 (58%) 0 (0%) 0.002
SV T/V F, n (%) 3 (50%) 0 (0%) 2 (33%) 0 (0%) 5 (26%) 0 (0%) 0.129
Indexed LVEDV (ml/m2)74 ± 16 66 ± 12 90 ± 7 86 ± 26 78 ± 17
LVEDD (mm) 52 ± 6 48± 4 53 ±953 ± 5 52 ± 6 44 ± 3 <0.001
LVEF (%) 45 ± 13 53 ± 6 41 ± 15 56 ± 5 47 ± 12 59 ± 3 0.001
CMRI, n (%) 5 (83%) 2 (50%) 3 (50%) 2 (66%) 12 (63%) /
LGE on CMRI, n (%) 4 (80%) 0 (0%) 1 (33%) 0 (0%) 5 (42%) /
PM/ICD implantation, n (%) 5 (83%) 2 (50%) 2 (33%) 1 (33%) 9 (47%) /
Heart transplant, n (%) 1 (17%) 0 (0%) 0 (0%) 0 (0%) 1 (5%) /
Note: Mean ± Standard Deviation.
Abbreviations: AF, atrial fibrillation; AVB, atrioventricular block; BMI, body mass index; CMRI , cardiac magnetic resonance imaging; DBP, diastolic blood pressure; DCM, dilated cardiomyopathy; ICD,
implantable cardioverter- defibrillator; LGE, late gadolinium enhancement; LVEDD, left ventricular end- diastolic diameter; LVEDV, left ventricular end- diastolic volume; LVEF, left ventricular ejection fraction;
NSVT, non- sustained ventricular tachycardia; PM, pacemaker; PVCs, premature ventricular complexes; SBP, systolic blood pressure; SVT, sust ained ventricular tachyc ardia; VF, ventricular fibrillation.
p- values ≤ 0.05 were considered statistically significant and repor ted in bold.
10906 
|
    G ERBINO E t al.
detected in three of them (Figure 2D). Finally, a patient of this group
underwent heart transplant (after unsuccessful VT trans- catheter ab-
lation) at the age of 45 due to sustained VT recurrences in storm and
subsequent LV function deterioration (LEVF 30%).17
3.2.2  |  Family 2
The patients of this family, with the p. Arg110Leufs*7 LMNA patho-
genic variant, showed a less aggressive clinical phenotype in com-
parison with members of the previously described family. In these
subjects, ventricular dimensions were in the normal range (LVEDD
48 ± 4 mm; LVEDVi 66 ± 12 ml/m2) and only mild left ventricular
systolic dysfunction was observed (LVEF 53 ± 6%). Half of the car-
riers developed conduction disturbances (first- degree AV block),
atrial fibrillation and ventricular arrhythmias as NSVT episodes,
and many PVCs were recorded at the 24- hour Holter monitoring.
Two subjects underwent ICD implantation in primar y prevention,
and one of them, with moderate left ventricular systolic dysfunc-
tion (LVEF 45%), underwent up- grading of dual- chamber pacemaker
to biventricular ICD with subsequent recovery of the left ventricu-
lar systolic function. To date, no members of this family presented
life- threatening ventricular tachyarrhythmias. Furthermore, two
patients carrying the above variant showed neuromuscular involve-
ment (radiculopathy associated with motor block, waddling gait and
girdle hypertrophy) still undergoing diagnostic definition.
FIGURE 1 Cardiac phenotype
evaluated by Echocardiographic imaging
in LMNA mutants’ carriers. The upper
half of the image shows LMNA DCM
phenotype: (A) and (B) panels show apical
four chamber (4C) views with chamber
dilatation (LVEDVi 82 ml/m2 and LVESVi
52 ml/m2) and reduced left ventricular
systolic function (LVEF 35%) due to
global hypocontractility. (C) PLA X view:
LVEDD (55 mm). (D) PSAX view: mid- LV
at the level of papillar y muscles. The
lower part (panels E– H) of the picture
shows a hypokinetic non- dilated LMNA
cardiomyopathy characterized by normal
ventricular volumes both in end- diastole
(E) (LVEDVi 64 ml/m2) and end- systole
(F) (LVESVi 38.8 ml/m2) with moderate
systolic left ventricular dysfunction
(LVEF 36%). (G) In PLAX view: LVEDD
(51 mm). (H) PSAX view: mid- LV at the
level of papillary muscles. DCM, dilated
cardiomyopathy; LVEDD, left ventricular
end- diastolic diameter; LVEDV, left
ventricular end- diastolic volume index;
LVEF, left ventricular ejection fraction;
LVESVi, left ventricular end- systolic
volume index; PLAX view, parasternal
long- axis view; PSAX, parasternal short-
axis view
   
|
10907
GERBIN O Et al.
FIGURE 2 Abnormal ECG findings in LMNA mutants’ carriers. (A) Episode of non- sustained VT with LBBB morphology and inferior
axis on 12- lead Holter monitoring. (B) ECG showing sinus rhythm, first- degree atrioventricular block (upper trace), type 1 s- degree
atrioventricular block (middle trace) and 2:1 atrioventricular block (lower trace) recorded in the same patient during 24- h ECG recording. (C)
12- lead ECG displaying atrial fibrillation and ventricular rhythm induced by pacemaker with VVI pacing mode. (D) ICD remote monitoring
report showing PVCs which trigger an episode of sustained TV, terminated, after ineffective ATP, by internal ICD shock. ATP, antitachycardia
pacing; ICD, implantable cardioverter- defibrillator; LBBB, left bundle branch block; PVCs, premature ventricular complexes; VT, ventricular
tachycardia
10908 
|
    G ERBINO E t al.
3.2.3  |  Family 3
Cardiac phenotype of p. Glu317Lys LMNA variant carriers was
characterized by left ventricular dilatation (LVEDD 53 ± 9 mm;
LVEDVi 90 ± 7 ml/m2) and systolic dysfunction (LVEF 41 ± 15%).
Three patients also displayed conduction disorders: 2 of them
presented first- degree AV- blocks with very long PR inter vals (the
longest reported was 520 ms), while the third patient developed
high- degree AV block and received a dual- chamber pacemaker sub-
sequently upgraded to a biventricular defibrillator. Atrial fibrillation
and NSV T were found in 50% of these patients, while 2 patients
also presented with SV T (one of them occurring during exercise
testing, and the other was detected on defibrillator arrhythmia
registr y). One patient received an ICD in primar y prevention due
to severe left ventricular dysfunction (LVEF 20%), which subse-
quently progre ssively imp roved (last reported LVEF was 47%) af ter
optimized drug treatment for heart failure, and the occurrence of
several appropriate defibrillator inter ventions by ATP on SVT was
also reported.
3.2.4  |  Family 4
The p. Arg190Gln LMNA variant was related to neuromuscu-
lar involvement consisting of muscle cramps, reduced strength to
stress and inappropriate muscular hypertrophy, associated with
significantly increased CPK values (1274 ± 1255 U/L), in addition
to cardiac abnormalities. One of the variant carriers had left ven-
tricular dilatation with normal ventricular function, while his brother
showed a hypokinetic non- dilated cardiomyopathy phenotype with
mildly impaired left ventricular systolic function (LVEF 50%) and nor-
mal ventricular volumes (Figure 1E– H); thus far, none of these two
patients has developed ventricular and supraventricular arrhyth-
mias or conduction disturbances. The third member of this family,
the father, having normal left ventricular size and systolic function,
showed paroxysmal atrial fibrillation with low ventricular rate and
underwent ICD implantation in primary prevention due to sinus
node disease and a wide QRS tachycardia episode occurrence dur-
ing exercise testing.
All lmna mutation- negative family members were clinically
asymptomatic, and none of them showed LMNA- linked cardiac
phenotypes either in terms of electrical disorders or mechanical ab-
normalities (Table 1).
3.3  | Cytokine and chemokine levels in the
sera of the LMNA patients and comparison with
those of healthy controls
Sera from the patients and controls described in the Table 2 were
screened for the circulating levels of 30 cytokines/chemokines
(Table 2). The levels of IL- 15 are under the lower limit of the assay
sensitivity, thus resulted undetectable in our cohort of patients. This
is in line with the fact that it has been reported that in humans, the
levels of circulating IL- 15 under normal conditions are low or unde-
tectable (~1 pg/ml ). 22
The levels of several pro- inflammatory cy tokines resulted up-
regulated in the sera of patients compared with controls. A more
detailed analysis of these cytokines was performed.
As shown in Figure 3A and Table 2, IL- 1ra resulted signifi-
cantly upregulated in the serum of both p. Leu140_Ala146dup, p.
Arg110Leufs*7 and p. Glu317Lys carriers compared with controls
(p = 0.0001, p = 0.0016 and p = 0.0002 respectively). The area
under the ROC curve (AUC) for IL- 1ra was 1 (Figure 3A’, p = 0.0003),
suggesting that the measurement of the levels of this cytokine
might allow us to discriminate between patients carrying either p.
Leu140_Ala146dup, p. Arg110Leufs*7 or p. Glu317Lys LMNA vari-
ants and controls enrolled in our study with high degree of accuracy.
Moreover, levels of IL- 8 resulted significantly increased in the serum
of p. Arg110Leufs*7, p. Glu317Lys and p. Arg190Gln LMNA carri-
ers compared with controls (Figure 3B, Table 2). The AUC values are
0.93 ± 0.04 (Figure 3B’, p = 0.0 004), indicating that IL- 8 levels may
be also considered as accurate biomarkers for the cardiomyopathy in
p. Arg110Leufs*7, p. Glu317Lys and p. Arg190Gln carriers.
Interestingly, the levels of both IL- 6 and G- CSF were signifi-
cantly increased in all patients compared with controls (Figure 4A , B,
Table 2) with the AUC value of 0.94 ± 0.04 (Figure 4A’, p = 0.0003)
and 1 (Figure 4B’, p < 0.00 01), respectively, indicating the levels of
these cytokines as valuable parameters to distinguish between pa-
tients af fected by LMNA- associated cardiomyopathy and controls at
least in our cohort of 30 individuals analysed.
Serum levels of IL- 1β, IL- 4, IL- 5 and GM- CSF were also signifi-
cantly upregulated in some of the families enrolled in the study
compared with controls (Figure 5). Interestingly, these cytokines
were more significantly upregulated in p. Leu140_Ala146dup car-
riers sug gesting a more complex pattern of inflammation in these
patients. Interestingly, amongst the cohort of p. Leu140_ Ala146dup
carriers, we identified a 57- year- old patient who underwent heart
transplantation in 2017 and asymptomatic at the time of the analy-
sis. In this carrier, the serum levels of cytokines were comparable to
that of control healthy subject s, thus suggesting a clear correlation
between the clinical manifestation of the cardiomyopathy and the
increased levels of pro- inflammatory cytokines (not shown).
3.4  | Analysis of Hsp70 in serum exosomes of
LMNA mutant carriers
It has been reported that extracellular Hsp70 may act through sur-
face receptors stimulating release of pro- inflammatory cytokines23
and that elevated serum levels of Hsp70 correlate with hypertrophy
and fibrosis in cardiovascular diseases.24
Indeed, to investigate more in deep the molecular mechanisms
involved in the establishment of the inflammatory phenot ype in
LMNA mutant carriers, we analysed the expression of Hsp70 in
serum exosomes of both patients and controls involved in the study.
   
|
10909
GERBIN O Et al.
TABLE 2 Serum levels of chemokines/cytokines expressed in pg/ml. Only the p values of significative differences compared with controls
were indicated (in bold). OOR< indicates that cytokine levels are under the lower limit of the assay sensitivity
p. Leu140_Ala146dup (n = 6) p. Arg110Leufs*7 (n = 4) p. Glu317Lys (n = 6) p. A rg190Gln (n = 3) Controls (n = 11)
IL- 1β6. 530 ± 1.522
p = 0.0011
3.715 ± 1.937
p = 0.56
4.918 ± 1.3 61
p = 0.01
2.643 ± 0.6233
p = 0.5
1.849 ± 0.2817
IL- 1r a 398.4 ± 30.03
p = 0.001
279.5 ± 14.29
p = 0.0016
396.0 ± 39. 83
p = 0.00 02
213.9 ± 82. 23
p = 0.22
132.4 ± 23 .15
IL- 2 9. 259 ± 2.24 4
p = 0.5608
8.193 ± 2.0 69
p = 0.7
12.95 ± 4.312
p = 0.19
8.237 ± 1 .411
p = 0.74
7.6 98 ± 0. 8279
IL- 4 9.68 0 ± 1.680
p = 0.00 05
4.578 ± 1.091
p = 0.52
7.9 4 8 ± 3.129
p = 0.16
4.523 ± 0.8 520
p = 0.53
3.343 ± 0.4856
IL- 5 20.99 ± 4.10 0
p = 0.00 04
9.923 ± 1.691
p = 0.07
31.84 ± 11.3 2
p = 0.0064
18.76 ± 10 .69
p = 0.05
5.679 ± 1.150
I L - 6 5.82 ± 1.0
p = 0.00 4
7.195 ± 1.805
p = 0.003
11.79 ± 3.19
p = 0.001
6.657 ± 1.76 4
p = 0.00 4
2.743 ± 0.3996
I L - 7 3 9.71 ± 5.560
p = 0.1554
29.17 ± 4.482
p = 0.78
35.56 ± 3. 523
p = 0.31
30.71 ± 8.330
p = 0.99
30.68 ± 2 .903
IL- 8 75.39 ± 18.43
p = 0.7383
198.8 ± 35.50
p = 0.005
256.5 ± 41 .4 6
p = 0.00 03
216.5 ± 36.85
p = 0.003
83.73 ± 16. 44
IL- 9 140.8 ± 6.28 8
p = 0.4683
139.1 ± 6.032
p = 0.58
169. 2 ± 14.11
p = 0.44
141.0 ± 11. 05
p = 0.68
152.4 ± 13.69
IL- 10 2.684 ± 0.6863
p = 0.9671
1.677 ± 0.7888
p = 0.49
2.564 ± 1.309
p = 0.97
2.523 ± 0.658 4
p = 0.93
2.633 ± 1.020
I L - 1 2 ( p 7 0 ) 6.952 ± 0.9793
p = 0.5925
6.720 ± 1. 217
p = 0.7
9.2 6 3 ± 1.709
p = 0.07
11.0 6 ± 4.952
p = 0.10
6.298 ± 0.7212
I L - 1 3 4.295 ± 0.8268
p = 0.6949
3.093 ± 0.3060
p = 0.67
5.575 ± 0.9616
p = 0.24
3.083 ± 0.8539
p = 0.72
3.785 ± 0.9604
IL- 15 OOR<OOR<OOR<OOR<OOR<
I L - 1 7 A 2 7.16 ± 5 .176
p = 0.186 0
24.26 ± 6.430
p = 0.34
39. 71 ± 14. 88
p = 0.09
24.65 ± 3.60 0
p = 0.21
20.02 ± 1.597
Eotaxin 1 7 7. 8 ± 29. 02
p = 0.3068
138.6 ± 21.04
p = 0.94
178.0 ± 22.04
p = 0.26
130.9 ± 38.26
p = 0.81
141. 2 ± 20. 28
bFGF 54.50 ± 6.378
p = 0.3689
52.57 ± 7.433
p = 0.49
72.84 ± 16 .73
p = 0.08
56.65 ± 6.106
p = 0.19
48.02 ± 2 .953
G - C S F 2980 ± 523.8
p = 0.00 01
1486 ± 442 .6
p = 0.0035
1140 ± 220.4
p = 0.002
1398 ± 188.5
p = 0.00 01
340.6 ± 57. 89
G M - C S F 6.250 ± 1.060
p = 0.00 05
3.067 ± 1.011
p = 0.19
5.355 ± 0.9873
p = 0.05
5.473 ± 2.787
p = 0.06
1.978 ± 0.3146
IFN- γ14.97± 5.036 10.3 3±0.8 819 11. 35± 1.888 12.45± 5.019 11.47± 4.499
IP - 10 748.8 ± 52.22
p = 0.0650
610. 3 ± 105.9
p = 0.84
1030 ± 285.7
p = 0.08
402.4 ± 53 .65
p = 0.13
58 7.4 ± 63.58
MCP- 1 (MCAF) 37.97 ± 4.896
p = 0.1871
31.78 ± 3.089
p = 0.10
73.28 ± 15.05
p = 0.11
37. 91 ± 5.729
p = 0.35
48.88 ± 6.413
MIP- 1α21.52 ± 8.44 3
p = 0.4025
33. 59 ± 14. 43
p = 0.08
29. 0 8 ± 6 .912
p = 0.06
40.18 ± 1.161
p = 0.005
14.02 ± 3.919
PDGF- bb 3233 ± 360.7
p = 0.2717
3388 ± 1 77. 8
p = 0.52
38 51 ± 394.6
p = 0.96
4797 ± 332.4
p = 0.31
3883 ± 4 37.6
MIP- 1b 148.5 ± 35.34
p = 0. 3511
142. 2 ± 23. 39
p = 0.28
155.5 ± 22.78
p = 0.10
108. 4 ± 10. 58
p = 0.82
114.0 ± 12. 52
RANTES 11929 ± 556 .9
p = 0.1784
1244 6 ± 421.6
p = 0. 51
14188 ± 518 .9
p = 0.711
11606 ± 1082
p = 0.35
13630 ± 1043
TNF- γ99. 09 ± 2 9.64
p = 0.2234
80.49 ± 21.18
p = 0.23
95.85 ± 20.49
p = 0.05
64.04 ± 7.89 3
p = 0.92
63.16 ± 4.318
(Continues)
10910 
|
    G ERBINO E t al.
We found an increase in the Hsp70 expression in serum exo-
somes from LMNA- p. Leu140_Ala146dup carriers compared with
controls (Figure 6A). The same exosomes were tested for the ex-
pression of the exosome’ markers, CD81 and CD9 (Figure 6B).
Densitometric analysis of Hsp70 band in the serum of all patients
and controls showed that only the circulating levels of Hsp70 were
significantly upregulated in LMNA- p. Leu140_Ala146dup carriers
compared with controls (Figure 6C).
To corroborate the relationship bet ween the elevated serum lev-
els of Hsp70 in serum exosomes from LMNA- p. Leu140_ Ala146dup
carriers and the specific LMNA mut ant, we analysed the expression
levels of Hsp70 in HEK293 cells after LMNA- p. Leu140_Ala146dup
expression. As shown in Figure 6D and E, the expression levels of
Hsp70 increased by about twofold in LMNA- p. Leu140_Ala146dup
compared with LMNA- WT- expressing HEK293 cells.
4 | DISCUSSION
In this work, we found that specific pro- inflammatory cytokines re-
sulted upregulated in a cohort of patients affec ted by cardiomyopa-
thy due to different mutations in lmna gene.
Carriers expressing the pathogenic LMNA variant are character-
ized by bradycardia, AF, a signific ant increase in PR interval and QRS
duration, high frequency of AV block, PVCs and NSVT occurrence,
an increase in LVEDD and a decrease in LVEF compared with con-
trols, respectively (Table 1).
The PR inter val duration >200 ms and a QRS complex prolonga-
tion (Table 1) denote the presence of AV- conduction blocks and an
impaired electrical conduction within the ventricles in LMNA variant
ca rri ers . In ad d it ion , hig h fre que ncy of PVC s an d NSVT in LMNA mu-
tant carriers clearly demonstrates the presence of early depolariza-
tions originating in the ventricles of these patients due to increased
ventricular automaticity. Both PVCs and NSVT are associated with
an overall increased risk for clinically relevant heart failure and an in-
creased risk for death.25 Moreover, LMNA mutant carries recruited
in this study are charac terized by mean increase in LVEDD and a
decrease in LVEF compared with controls denoting a left ventricular
dilation and dysfunction.
We found high levels of circulating G- CSF and IL- 6 in all patient s,
and elevated levels of IL- 1ra and IL- 8 in a large subset of the patients
enrolled in the study.
In particular, high levels of both G- C SF and IL- 6 were obser ved in
all the families we examined, independently from the mutation t ype
and the phenotypes detected in each family. On the base of these
findings, it is possible to hypothesize that high levels of both G- CSF
and IL- 6 could be associated with cardiolaminopathies. On the con-
trary, high values of IL- 1ra are present in three (Families 1, 2 and 3) of
the investigated families whose members carrying the LMNA variant
displayed a high percent age of AV- blocks, NSVT and PVCs, which
are completely absent from patients of the fourth family and from
control subjects (Table 1). In addition to this, the observed PR in-
tervals were significantly longer in patients from Families 1, 2 and 3
when compared either to patients from the fourth family or subjec ts
not carr ying LMNA variants (Table 1). Interestingly, the members of
Family 4 were characterized by a different laminopathic phenotype
than those belonging to the other three families, because they dis-
played a clear neuromuscular involvement associated with high CPK
values and less severe electrical and mechanical cardiac damage.
This suggests that IL- 1ra could be a biomarker associated with con-
duction defects and arrhythmic manifestations in LMNA patients
carrying an over t cardiac phenotype.
G- CSF is produced by bone marrow stromal cells, endothelial
cells, macrophages and fibroblasts. Its production is induced by in-
flammatory stimuli such as pro- inflammatory cytokines (TNF- α, I L - 6
and IL- 1), and it may enhance the pro- inflammatory responses by
controlling neutrophil numbers and their activity during inflamma-
tion. Although we did not find any signific ant increase in TNF- α in
LMNA mutant carriers, we did measure a significant increase in the
serum levels of IL- 1ra in p. Leu140_Ala146dup, p. Arg110Leufs*7
and p. Glu317 Lys ca rr iers an d in th e se rum level s of IL- 6 in all LMNA
mutants carriers compared with controls. IL- 1ra is a receptor an-
tagonist of IL- 1 activity, and it is released rapidly in the circulation
under the same inflammator y conditions that stimulate IL- 1α and
p. Leu140_Ala146dup (n = 6) p. Arg110Leufs*7 (n = 4) p. Glu317Lys (n = 6) p. A rg190Gln (n = 3) Controls (n = 11)
VEGF 108.2 ± 47.78
p = 0.555 4
20 9.5 ± 86 .52
p = 0.15
133.4 ± 47.72
p = 0.34
161. 0 ± 43.49
p = 0.14
72.98 ± 29. 99
TGF- β149020 ± 2220
p = 0.6832
54484 ± 476 .7
p = 0.9888
51638 ± 4105
p = 0.9959
63726 ± 3358
p = 0.1202
52788 ± 2577
TGF- β23169 ± 75. 88
p = 0.9999
3282 ± 65.51
p = 0.8934
3421 ± 172.1
p = 0.196 8
3244 ± 186.3
p = 0.9827
3173 ± 35 .17
TGF- β31627 ± 23.58
p = 0.6128
1666 ± 57.1 2
p = 0.9790
1548 ± 105,3
p = 0.153 4
1770 ± 83.44
p = 0.920 4
1705 ± 38.60
Abbreviations: IL, interleukin; bFGF, basic fibroblas t growth factor; G- CSF, granulocyte colony- stimulating f actor; GM- CSF, granuloc yte- macrophage
colony- stimulating factor; IFN- γ, γ- interferon; IP- 10, interferon gamma- induced protein- 10; MCP- 1, monocyte chemoattractant protein- 1; MIP- 1α,
MIP- 1β, macrophage inflammatory protein- 1α and 1β; PDGF- BB, platelet- derived grow th fac tor- BB; TNF- α, tumour necrosis factor- α; VEGF, vascular
endothelial growth factor; TGF- β, transforming growth factor- β.
p- values ≤ 0.05 were considered statistically significant and repor ted in bold.
TABLE 2 (Continued)
   
|
10911
GERBIN O Et al.
IL- 1β. Of note, the measurement of IL- 1ra levels rather than IL- 1α
or IL- 1β is a more reliable parameter of an increase in production
of IL- 1 family members in inflammator y conditions since IL- 1α and
IL- 1β lack the secretory peptide signal, and thus, they are not readily
secreted into the systemic circulation.
IL- 1 family members can act on cardiac resident macrophages,
neutrophils and parenchymal cells to trigger production of IL- 6, G-
CSF and other inflammator y mediators. It has been reported that IL- 1
is consistently upregulated in experimental models of heart failure
due to a wide range of aetiologies, including myocardial infarc tion
and diabetic cardiomyopathy.26 Moreover, there are experimental
evidence supporting the role of IL- 1 signalling in the pathogenesis
of cardiac dysfunction and adverse remodelling associated with
heart failure. For instance, KO mice for IL- 1 receptors showed at-
tenuated adverse remodelling after myocardial infarction, exhibit-
ing suppressed inflammatory responses.27 IL- 1 suppresses systolic
FIGURE 3 Serum levels of IL1- ra
(A) and IL- 8 (B) in the family enrolled in
the study and in healthy controls. **p <
.01; ***p < .001. Areas under the curves
(AUC) obtained with the sensitivity and
specificity for the serum levels of IL1- ra
(A’) and IL- 8 (B’) in patients with LMNA
mutant shown in A and in B respectively
FIGURE 4 Serum levels of IL6 (A) and
G- CSF (B) in the family enrolled in the
study and in healthy controls. **p < .01;
***p < .001; ****p < .0001. Areas under
the curves (AUC) obtained with the
sensitivity and specificity for the serum
levels of IL6 (A’) and G- CSF (B’) in patients
with LMNA mutant shown in A and in B,
respectively
10912 
|
    G ERBINO E t al.
cardiomyocyte function by the disruption of calcium handling28
and by promulgating cardiomyocyte apoptosis.29 In agreement with
these experimental evidence, our data support the above pathoge-
netic mechanisms because we found high levels of IL- 1β in Families 1
and 3. In particular, these patients displayed signific antly lower LVEF
values and higher prevalence of serious arrhythmic events in com-
parison with patients from other families (Table 1). Both left ventric-
ular dysfunction and arrhythmias seem to be associated with IL- 1β
increased levels in our patients, thus potentially confirming the role
of this cytokine as a biomarker related to more severe cardiac me-
chanica l and electrical abnormalities in LMNA patients. Interestingly,
IL- 1ra and G- CSF levels were found upregulated in patient cohort
affected by striated muscle laminopathies, including LMNA cardio-
myopathy,30 thus suggesting not only a link of these cytokines with
cardiac dysfunction but also laminopathies in general.
The role of IL- 6 in the pathogenesis of cardiac disease is well es-
tablished in experimental models and in humans. IL- 6 is consistently
upregulated in experimental models of cardiac injury and hear t
failure regardless of the underlying aetiology and is expressed by
cardiomyocytes, infiltrating mononuclear cells and fibroblasts.31,32 In
cardiomyocytes, IL- 6 is able to decrease intracellular Ca2+ transients
and depress cell contraction through a nitric oxide (NO)- cGMP-
mediated pathway,33,34 and in fibroblasts, IL- 6 promotes prolifera-
tion and stimulates extracellular matrix synthesis.34 Moreover, in in
vivo studies, infusion of IL- 6 caused hyper trophy and fibrosis, and
increased myocardial stiffness in mice.35
Importantly, high plasma levels of IL- 6 can provide prognostic in-
formation in patient s with chronic heart failure (CHF), independently
of ventricular dysfunction and of aetiology, suggesting an important
role for IL- 6 in the pathophysiology of HF.36,37 Regardless of specific
aetiology and organ localization, systemic inflammation, via IL- 6 eleva-
tion, rapidly induces atrial electrical remodelling and electrical instabil-
ity thus increasing susceptibility to atrial fibrillation, by downregulating
cardiac connexins.38 Emerging experimental evidence suggests that
IL- 6 may play a critical role in contributing to the modulation of ICa,
L and IK current s, and both factors are active contributors to cardiac
FIGURE 5 Serum levels of IL- 1β, IL- 4,
IL- 5 and GM- CSF in some of the family
enrolled in the study and in healthy
controls. **p < .01; ***p < .001
   
|
10913
GERBIN O Et al.
instabilities.39 In accordance with these experimental findings, atrial
fibrill ation occurrence w as documented in all e valuated LMNA fami lies.
IL- 8, which also resulted upregulated in most of the patients in-
volved in this study, has been shown to be induced in the failing myo-
cardium,40 and it was reported to predict the development of left
ventricular dysfunction and the following HF together with IL- 6.41
Interestingly, Family 4 with a neuromuscular phenotype shows
circulating levels MIP- 1α significantly higher than those from other
family patients or control subject s. The association between high ex-
pression of this biomarker and neuromuscular phenotype in LMNA
variant patients is consistent with data from Cappelletti C et al. who
examined the cytokine profile in patients with striated muscle lami-
nopathies.30 Thus, MIP- 1α could be associated with skeletal muscle
involvement in cardiac laminopathy patients.
In addition to the cy tokines discussed so far, IL- 4, IL- 5 and GM-
CSF resulted also significantly increased in the serum of some pa-
tients enrolled in the study.
A recent and elegant study demonstrated the cooperative role of
IL- 5 and IL- 4 in the development of inflammatory dilated cardiomy-
opathy (DCMi). Transgenic mice overexpressing IL- 5 developed eo-
sinophil infiltration and a severe spontaneous cardiac enlargement.
The role of IL- 5 in the pathogenesis of DCMi was to mobilize IL- 4-
producing eosinophils into the myocardium, which then in turn were
responsible for the dilated cardiomyopathy.42 Accordingly, several
studies have shown a positive correlation between systemic IL- 4 lev-
els and cardiac fibrotic remodelling and dilation in both patients and
experimental animals.43- 45
Interestingly, p. Leu140_Ala146dup carriers with the widest
spectrum of pro- inflammatory circulating cytokines dysregulated
show the most severe cardiac phenotype in our cohort of LMNA
mutant carriers. Notably, IL- 4 and GM- CSF resulted to be highly ex-
pressed only in patients belonging to the above family. Since this
family displayed the most severe cardiac phenotype, both in terms
of cardiac function impairment and in relationship with malignant
arrhythmic event s occurrence, this further supports the hypothesis
that the pro- inflammatory cy tokines we found upregulated in our
study, significantly contribute to pathogenesis of the LMNA cardio-
myopathy at least in our cohort of LMNA mutant carriers.
To gain more insight s on the molecular events involved in the
inflammatory response in our patient s, we paid attention to the heat
shock proteins since some of them have been shown to be potent
activators of the innate immune system.46
Of note, we found significantly elevated expression levels of
Hsp70 in the serum exosomes of LMNA- p. Leu140_Ala146dup car-
riers and a significant increase in Hsp70 upregulation in LMNA- p.
Leu140_Ala146dup- expressing cells. It has been repor ted that
Hsp70 is released from cardiomyocytes undergoing lysis, necrosis
and apoptosis, as well as via active secretion in response to a vari-
ety of stress stimuli, including ischaemia and oxidative stress.47, 48
Accordingly, we previously reported that LMNA- p. Leu140_
Ala146dup- expressing cardiomyocytes have decreased nuclear
stability, resulting in a higher rate of apoptosis.17 High levels of cir-
culating Hps70 have been detec ted in patients with acute myocar-
dial infarction correlating with the extent of myoc ardial damage.49
Moreover, GM- CSF has been reported to significantly increases the
expression of Hsp70 in infarcted myocardium in mice.50 Indeed, the
most severe cardiac phenotype and the upregulated levels of GM-
CSF found only in LMNA- p. Leu140_Ala146dup carriers may ac-
count for the elevated levels of circulating Hsp70 in these patient s.
Of note, Hsp70 and IL- 6 are potential new therapeutic targets for
this subset of cardiolaminopathies.
In fact, a significant cardioprotective effect of the anti- Hsp70
blocking antibody has been demonstrated in an HF mouse model,
with resolution of myocardial inflammation, left ventricular dilation
FIGURE 6 Hsp70 expression in patients’ serum exosomes and in LMNA- p. Leu140_Ala146dup- expressing cells. (A) Western blot ting on
serum exosomes from LMNA- p. Leu140_Ala146dup- carrying patients (p. Leu140) and healthy controls (controls) with anti- Hsp70 antibodies.
Equal volumes of sera from each patient were loaded. (B) Western blotting on serum exosomes from LMNA- p. Leu140_Ala146dup- carrying
patients (p. Leu140) and healthy controls (controls) with anti- CD81, anti- CD- 9 antibodies used as markers of exosomes purity. GAPDH
absence is also index of exosome purity and was included in the panel. (C) Densitometric analysis of Hsp70- immunoreactive bands in serum
exosomes from all patients and controls included in the study. The data are means of 3 independent experiments. *p <  .05. (D) Western
blotting of Hsp70 and LMNA in either LMNA WT or LMNA- p. Leu140_Ala146dup- expressing HEK293 cells. (E) Densitometric analysis
of Hsp70- immunoreactive bands in either LMNA W T or LMNA- p. Leu140_ Ala146dup- expressing HEK293 cells. The data are means of 3
independent experiments. **p < .01
10914 
|
    G ERBINO E t al.
and dysfunction and a significant inhibition of cardiac fibrosis.51
Moreover, Hsp70 blocking improves cardiac functional recovery in
mice after global ischaemia reperfusion and reduces expression of
the pro- inflammatory cytokines TNF- α, IL- 1β and IL- 6.52 In the clin-
ical practice, an anti- IL6 receptor antibody (tocilizumab) is widely
used to treat the abnormal inflammator y response that occurs in
autoimmune diseases as rheumatoid arthritis and it is well tolerated
by patients.53 In addition, its in vivo administration to the mouse
model of progeria significantly ameliorates the progeroid pheno-
type, including cardiac histology in these mice.54 Of note, the anti-
IL- 6 receptor antibody (MR16- 1) prevents the development of LV
remodelling after MI in mice.55
5 | CONCLUSIONS
Clinical course of cardiac laminopathies is characterized by a poor
prognosis and a high rate of major cardiac events. So far, therapeutic
approaches are exclusively symptomatic. Improvement in therapeu-
tic management might come from ver y early treatment with drugs
hopefully already used in clinical practice. In this scenario, we be-
lieve that our data are of great interest on the translational point of
view. The main finding of our study is that inflammatory cytokines
could significantly contribute to the pathogenesis of the cardiomyo-
pathy in lmna mutation carriers and correlate with the severity of the
cardiac phenotype. Indeed, early identification of dysregulated pro-
inflammatory serum cytokines in LMNA- cardiomyopathy patients
could be crucial for therapeutic approaches able to counteract the
progression of the disease and to finally improve the prognosis of
this subset of severe cardiomyopathies.
ACKNOWLEDGEMENT
This work wa s sup por te d by fu ndi ng fro m ‘C arm osin o19LAMIN OPATIE’
and ‘Carmosino20RIL’ to Monica Carmosino and from the CLUSTER
TECNOLOGICO REGIONALE ‘DICLIMAX’ (project # MTJU9H8) to
Maria Svelto.
CONFLICT OF INTEREST
The authors have no conflict of interest to declare.
AUTHOR CONTRIBUTIONS
Andrea Gerbino: Conceptualization (equal); Data curation (equal);
Investigation (equal); Writing- original draft (equal). Cinzia Forleo:
Data curation (equal); Methodology (equal); Resources (equal);
Writing- original draft (equal). Serena Milano: Data curation (equal);
Formal analysis (supporting); Investigation (equal). Francesca
Piccapane: Data curation (equal); Methodology (equal). Giuseppe
Procino: Writing- review & editing (equal). Martino Pepe: Resources
(equal). Mara Piccolo: Resources (equal). Piero Guida: Data curation
(lead). Nicoletta Resta: Data curation; Investigation (equal). Stefano
Favale: Writing- review & editing (equal). Maria Svelto: Funding ac-
quisition (lead); Writing- review & editing (equal). Monica Carmosino:
Conceptualization (equal); Formal analysis (supporting); Funding ac-
quisition (supporting); Project administration (lead).
DATA AVAIL ABI LIT Y STAT EME NT
The data that support the findings of this study are available from
the corresponding author upon reasonable request.
ORCID
Monica Carmosino https://orcid.org/0000-0001-7600-8816
REFERENCES
1. Crasto S, My I, Di Pasquale E. The broad spec trum of LMNA cardiac
diseases: from molecular mechanisms to clinical phenotype. Front
Physiol. 2020;11:1- 11.
2. Captur G, Arbustini E, B onne G, et al. Lamin and the heart. Heart.
2 0 1 8 ; 1 0 4 : 4 6 8 - 4 7 9 .
3. Carmosino M, Torretta S, Procino G, et al. Role of nuclear Lamin
A/C in cardiomyocy te functions. Biol Cell. 2014;106(10):346- 358 .
4. Gerbino A, Procino G, Svelto M, et al. Role of Lamin A /C gene mu-
tations in the signaling defects leading to cardiomyopathies. Fron t
Physiol. 2018;9:1356.
5. Muchir A, Pavlidis P, Decostre V, et al. Activation of MAPK path-
ways links LMNA mut ations to c ardiomyopathy in Emery- Dreif uss
muscular dystrophy. J Clin Invest. 20 07;117:1282- 1293.
6. Muchir A , Wu W, Choi JC, et al. Abnormal p38α mitogen- activated
protein kinase signaling in dilated cardiomyopathy caused by lamin
A/C gene mut ation. Hum Mol Genet. 2012;21:4325- 4333.
7. Carmosino M, Gerbino A , Schena G, et al. The expression of L amin
A mut ant R321X leads to endoplasmic reticulum stress with aber-
rant Ca2+ handling. J Cell Mol Med. 2016;20(11):2194- 2207.
8. Dridi H, Wu W, Reiken SR, et al. Ryanodine receptor remodeling in
cardiomyopathy and muscular dystrophy caused by lamin A/C gene
mutation. Hum Mol Genet. 2021;29:3919- 3934.
9. Morales Rodriguez B, Domínguez- Rodríguez A, Benitah JP, et al.
Activation of sarcolipin expression and altered calcium cycling in
LMNA cardiomyopathy. Biochem Biophys Reports. 2020;22:100767.
10. Gerbino A , Bottillo I, Milano S, et al. Functional Characterization of
a Novel Truncating Mutation in Lamin A/C Gene in a Family with
a Severe Cardiomyopathy with Conduction Defects. Cell Physiol
Biochem. 2017;44(4):1559- 1577.
11. Macquar t C, Jüt tner R , Morales Rodriguez B, et al. Microtubule cy-
toskeleton regulates Connexin 43 localization and cardiac conduc-
tion in cardiomyopathy caused by mutation in A- type lamins gene.
Hum Mol Genet. 2019;28:4043- 4052.
12. Salvarani N, Crasto S, Miragoli M, et al. The K219T- Lamin muta-
tion induces conduction defects through epigenetic inhibition of
SCN5A in human cardiac laminopathy. Nat Commun. 2019;10:2267.
13. van Tintelen JP, Tio RA, Kerstjens- Frederikse WS, et al. Severe
myocardial fibrosis caused by a deletion of the 5’ end of the lamin
A/C gene. J Am Coll Cardiol. 20 07;49:243 0- 2439.
14. Holmström M, KivisS, Heliö T, et al. Late gadolinium enhanced
cardiovascular magnetic resonance of lamin A/C gene muta-
tion related dilated cardiomyopathy. J Cardiovasc Magn Reson.
2011;13(30):1- 9.
15. Sutton MGSJ, Sharpe N. Left ventricular remodeling after myo-
cardial infarction: Pathophysiology and therapy. Circulation.
2000;101:2981- 2988.
16. Krejci J, Mlejnek D, Sochorova D, et al. Inflammatory c ardiomyop-
athy: A current view on the pathophysiology, diagnosis, and treat-
ment. Biomed Res I nt. 2016;2016:1- 11.
17. Forleo C, Carmosino M, Resta N, et al. Clinical and func tional char-
acterization of a novel mutation in lamin a/C gene in a multigener-
ational family with arrhythmogenic cardiac laminopathy. PLoS One.
2015;10(4):e0121723.
18. Arbustini E, Pilot to A, Repetto A , et al. Autosomal dominant dilated
cardiomyopathy with atrioventricular block: A lamin A /C defect-
related disease. J A m Coll Cardiol. 2002;39:981- 990.
   
|
10915
GERBIN O Et al.
19. Villa F, Maciag A, Spinelli CC, et al. A G613A missense in the
Hutchinson’s progeria lamin A/C gene causes a lone, autosomal
dominant atrioventricular block. Immun Ageing. 2014;11(19):1- 6.
20. Perrot A, Hussein S, Ruppert V, et al. Identification of mutational
hot spots in LMNA encoding lamin A/C in patients with familial di-
lated cardiomyopathy. Basic Res Cardiol. 2 0 09;10 4:9 0 - 9 9.
21. Parks SB, Kushner JD, Nauman D, et al. Lamin A/C mutation analy-
sis in a cohort of 324 unrelated patients with idiopathic or familial
dilated cardiomyopathy. Am Heart J. 200 8;156:161- 169.
22. Bergamaschi C , Bear J, Rosati M, et al. Circulating IL- 15 exists as
heterodimeric complex with soluble IL- 15Rα in human and mouse
serum. Blood. 2012;120:e1- e8.
23. Asea A, Rehli M, Kabingu E, et al. Novel Signal Transduction
Pathway Utilized by Extracellular HSP70. J Biol Chem. 2002;277:
15028- 15034.
24. Cai W- F, Zhang X- W, Yan H- M, et al. Intracellular or extracellular
heat shock protein 70 differentially regulates cardiac remodelling
in pressure overload mice. Cardiovasc Res. 2010; 88:140 - 149.
25. Marcus GM. Evaluation and Management of Premature Ventr icular
Complexes. Circulation. 20 20;141:1404 - 1418.
26. Hanna A, Frangogiannis NG. Inflammatory Cytokines and Chemokines
as Therapeutic Targets in Heart Failure. Cardiovasc Drugs Ther. 2020;
34:849- 863.
27. Bujak M, Dobaczewski M, Chatila K, et al. Interleukin- 1 receptor
type I signaling critically regulates infarct healing and cardiac re-
modeling. Am J Pathol. 2008;173:57- 67.
28. Kumar A, Thota V, Dee L, et al. Tumor necrosis factor α and in-
terleukin 1β are responsible for in vitro myocardial cell depres-
sion induced by human septic shock serum. J Exp Med. 1996;18 3:
949- 958.
29. Ing DJ, Zang J, Dzau VJ, et al. Modulation of c ytokine- induced car-
diac myocyte apoptosis by nitric oxide, Bak, and Bcl- x. Circ Res.
1999;84:21- 33.
30. Cappelletti C, Tramacere I, Cavalc ante P, et al. Cytokine Profile
in Striated Muscle Laminopat hies: New Promising Biomarkers for
Disease Prediction. Cells. 2020;9:1532.
31. Gwechenberger M, Mendoza LH, Youker KA , et al. C ardiac myo-
cytes produce interleukin- 6 in culture and in viable border zone of
reperfused infarctions. Circulation. 1999;99:546- 551.
32. Kubota T, Miyagishima M, Alvarez RJ, et al. Expression of proin-
flammatory cytokines in the failing human heart: Comparison of
recent- onset and end- stage congestive heart f ailure. J Hear Lung
Transplant. 2000 ;19:819- 8 24.
33. Kinugawa KI, Takahashi T, Kohmoto O, et al. Nitric oxide- mediated
effec ts of interleukin- 6 on [Ca2+](i) and cell contrac tion in cultured
chick ventricular myocytes. Circ Res. 1994;75:285- 295.
34. Leicht M, Briest W, Zimmer HG. Regulation of norepinephrine-
induced proliferation in cardiac fibroblasts by interleukin- 6 and
p42/p44 mitogen ac tivated protein kinase. Mol Cell Biochem.
2003;243:65- 72.
35. Meléndez GC , McLarty JL, Levick SP, et al. Interleukin 6 mediates
myocardial fibrosis, concentric hypert rophy, and diastolic dysfunc-
tion in rat s. Hypertension. 2010;56:225- 231.
36. De swal A, Petersen NJ, Feldman AM, et al. Cytokines and cyto-
kine receptors in advanced heart failure: An analysis of the cy-
tokine database from the Vesnarinone Trial (VEST). Circulation.
2001;103:2055- 2059.
37. Tsutamoto T, Hisanaga T, Wada A, et al. Interleukin- 6 spillover in
the peripheral circulation increases with the severity of heart f ail-
ure, and the high plas ma le vel of interl eukin- 6 is an imp or tant prog-
nostic predictor in patients with congestive heart failure. J Am Coll
Cardiol. 1998;31:391- 398.
38. Lazzerini PE, Laghi- Pasini F, Acampa M, et al. Systemic Inflammation
Rapidly Induces Reversible Atrial Electrical Remodeling: The Role
of Interleukin- 6– Mediated Changes in Connexin Expression. J Am
Heart Assoc. 2019;8 (16):1- 17.
39. Hagiwara Y, Miyoshi S, Fukuda K, et al. SHP2- mediated signaling
cascade through gp130 is essential for LIF- dependent ICaL, [Ca2+]
i transient, and APD increase in cardiomyocytes. J Mol Cell Cardiol.
2007;43:710 - 716.
40. Damås JK, Eiken HG, Øie E, et al. Myocardial expression of CC-
and CXC- chemokines and their receptors in human end- stage heart
failure. Cardiovasc Res. 2000;47:778- 787.
41. De Gennaro L, Brunetti ND, Montrone D, et al. Subacute inflamma-
tory ac tivation in subjects with acute coronar y syndrome and left
ventricular dysfunction. Inflammation. 2012;35:363- 370.
42. Diny NL, Baldeviano GC, Talor MV, et al. Eosinophil- derived IL- 4
drives progression of myocarditis to inflammatory dilated cardio-
myopathy. J Exp Med. 2017;214:943- 957.
43. Cieslik KA, Taffet GE, Carlson S, et al. Immune- inflammatory dysreg-
ulation modulates the incidence of progressive fibrosis and diastolic
stiffness in the aging heart. J Mol Cell Cardiol. 2011;50:248- 256.
44. Levick SP, McLart y JL, Murray DB, et al. Cardiac mast cells mediate
left ventricular fibrosis in the hypertensive rat heart. Hypertension.
200 9;53:1041- 1047.
45. Peng H, Sar war Z, Yang XP, et al. Profibrotic role for interleukin- 4
in cardiac remodeling and dysfunction. Hypertension. 2015;66:
582- 589.
46. Tsan MF, Gao B. Cytokine function of heat shock proteins. Am J
Physiol Cell Physiol. 2004;286:C739- C744.
47. Ranek MJ, Stachowski MJ, K irk JA, et al. The role of heat shock
proteins and co- chaperones in heart failure. Philos Trans R Soc B Biol
Sci. 2018;373 (1738):1- 17.
48. Lin L, Knowlton A A. Innate immunity and cardiomyocytes in isch-
emic heart disease. Life Sci. 2014;100:1- 8 .
49. Dybdahl B, Slørdahl SA , Waage A, et al. Myocardial ischaemia and
the inflammatory response: Release of heat shock protein 70 af ter
myocardial infarction. Heart. 2005;91:299- 304.
50. Naito K, Anzai T, Sugano Y, et al. Differential Effects of GM- CSF
and G - CSF on Infiltration of Dendritic Cells during Early Left
Ventricular Remodeling after Myocardial Infarction. J Immunol.
2008;181:5691- 5701.
51. Liu P, Bao HY, Jin CC, et al. Targeting extracellular heat shock pro-
tein 70 ameliorates doxorubicin- induced hear t failure through res-
olution of toll- like receptor 2– mediated myocardial inflammation.
J Am Heart Assoc. 2019;8(20):1- 11.
52. Zou N, Ao L, Cleveland JC, et al. Critical role of extracellular heat
shock cognate protein 70 in the myocardial inflammatory response
and cardiac dysfunction after global ischemia- reperfusion. Am J
Physiol Hear Circ Physiol. 2008;294(6):H2805- H2813.
53. Kang S, Tanaka T, Narazaki M, et al. Targeting Interleukin- 6 Signaling
in Clinic. Immunity. 2019;50:1007- 1023.
54. Squarzoni S, Schena E, Sabatelli P, et al. Interleukin- 6 neutraliza-
tion ameliorates symptoms in prematurely aged mice. A ging Cell.
2021;20.1- 17.
55. Kobara M, Noda K, Kitamura M, et al. Antibody against interleu-
kin- 6 receptor attenuates left ventricular remodelling after myo-
cardial infarction in mice. Cardiovasc Res. 2010;87:424- 430.
SUPPORTING INFORMATION
Additional supporting information may be found in the online ver-
sion of the ar ticle at the publisher’s website.
How to cite this article: Gerbino A, Forleo C, Milano S, et al.
Pro- inflammatory cytokines as emerging molecular
determinants in cardiolaminopathies. J Cell Mol Med.
2021;25:10902– 10915. https://doi. or g/10.1111/ jcmm.16975
... ; https://doi.org/10.1101/2024.06.11.598511 doi: bioRxiv preprint In contrast to fibroblasts, for which we did not detect an increased presence in Lmna-cKO hearts (Fig. 5B), our snRNA-seq analysis revealed an increase of immune cells in the Lmna-cKO hearts compared to the very low levels seen in the hearts of Lmna-cWT controls ( Figure 5C-D). These findings are consistent with previous LMNA-DCM studies 28,75 and reports that cardiomyocytes, in response to injury, release cytokines and recruit immune cells, leading to inflammation and further induction of cardiac remodeling 82,83 . Due to the low numbers of immune cells in the Lmna-cWT hearts at 25-dpi, determining disease-specific immune cell subclusters was challenging. ...
Preprint
LMNA-related dilated cardiomyopathy (LMNA-DCM) is one of the most severe forms of DCM. The incomplete understanding of the molecular disease mechanisms results in lacking treatment options, leading to high mortality amongst patients. Here, using an inducible, cardiomyocyte-specific lamin A/C depletion mouse model, we conducted a comprehensive transcriptomic study, combining both bulk and single nucleus RNA sequencing, and spanning LMNA-DCM disease progression, to identify potential disease drivers. Our refined analysis pipeline identified 496 genes already misregulated early in disease. The expression of these genes was largely driven by disease specific cardiomyocyte sub-populations and involved biological processes mediating cellular response to DNA damage, cytosolic pattern recognition, and innate immunity. Indeed, DNA damage in LMNA-DCM hearts was significantly increased early in disease and correlated with reduced cardiomyocyte lamin A levels. Activation of cytosolic pattern recognition in cardiomyocytes was independent of cGAS, which is rarely expressed in cardiomyocytes, but likely occurred downstream of other pattern recognition sensors such as IFI16. Altered gene expression in cardiac fibroblasts and immune cell infiltration further contributed to tissue-wide changes in gene expression. Our transcriptomic analysis further predicted significant alterations in cell-cell communication between cardiomyocytes, fibroblasts, and immune cells, mediated through early changes in the extracellular matrix (ECM) in the LMNA-DCM hearts. Taken together, our work suggests a model in which nuclear damage in cardiomyocytes leads to activation of DNA damage responses, cytosolic pattern recognition pathway, and other signaling pathways that activate inflammation, immune cell recruitment, and transcriptional changes in cardiac fibroblasts, which collectively drive LMNA-DCM pathogenesis.
... [62,72,94] Decreased WNT-β-catenin signaling related to pathogenic LMNA variants was shown in animal models. [95] Plasma cytokine profiles were shown to be different in LMNA cardiomyopathy with variable profiles in patients based on genotype/phenotype subtype providing a potentially inflammatory link to degree of disease manifestation driven by underlying variant [96]. Pathogenic LMNA variants are occasionally reported in case series of genetic testing in AM [9] but The presented proportions are based on a single study including only 36 patients [21]; confirmatory data are still required. ...
... Patients with laminopathy often exhibit increased peripheral inflammation, reflected by the higher levels of IL-1β, IL-4, and IL-6 in sera compared with those in healthy controls (Cappelletti et al., 2020;Gerbino et al., 2021). Furthermore, intrinsic aberrant activation in the immune system and systemic inflammation have been reported in animal models of progeroid syndromes (Osorio et al., 2012). ...
Article
Full-text available
Lamin A/C is involved in macrophage activation and premature aging, also known as progeria. As the resident macrophage in brain, overactivation of microglia causes brain inflammation, promoting aging and brain disease. In this study, we investigated the role of Lamin A/C in microglial activation and its impact on progeria using Lmna −/− mice, primary microglia, Lmna knockout ( Lmna ‐KO) and Lmna ‐knockdown ( Lmna ‐KD) BV2 cell lines. We found that the microglial activation signatures, including cell proliferation, morphology changes, and proinflammatory cytokine secretion (IL‐1β, IL‐6, and TNF‐α), were significantly suppressed in all Lamin A/C‐deficient models when stimulated with LPS. TMT‐based quantitative proteomic and bioinformatic analysis were further applied to explore the mechanism of Lamin A/C‐regulated microglia activation from the proteome level. The results revealed that immune response and phagocytosis were impaired in Lmna −/− microglia. Stat1 was identified as the hub protein in the mechanism by which Lamin A/C regulates microglial activation. Additionally, DNA replication, chromatin organization, and mRNA processing were also altered by Lamin A/C, with Ki67 fulfilling the main hub function. Lamin A/C is a mechanosensitive protein and, the immune‐ and proliferation‐related biological processes are also regulated by mechanotransduction. We speculate that Lamin A/C‐mediated mechanotransduction is required for microglial activation. Our study proposes a novel mechanism for microglial activation mediated by Lamin A/C.
... Activation of resident or infiltration of blood-derived immune cells are involved in pathological inflammatory pathways and tissue reparative processes in HF [55][56][57]. Upregulation of inflammatory cytokines contributes to the pathogenesis of LMNA-cardiomyopathy patients and affects the severity of cardiac phenotype [58]. Reduced immune cell numbers correlated with long term animal survival following treatment by Sun1 shRNA or Lamin C supplementation in Lmna DCM. ...
Article
Full-text available
Background Dilated cardiomyopathy (DCM) is a severe, non-ischemic heart disease which ultimately results in heart failure (HF). Decades of research on DCM have revealed diverse aetiologies. Among them, familial DCM is the major form of DCM, with pathogenic variants in LMNA being the second most common form of autosomal dominant DCM. LMNA DCM is a multifactorial and complex disease with no specific treatment thus far. Many studies have demonstrated that perturbing candidates related to various dysregulated pathways ameliorate LMNA DCM. However, it is unknown whether these candidates could serve as potential therapeutic targets especially in long term efficacy. Methods We evaluated 14 potential candidates including Lmna gene products (Lamin A and Lamin C), key signaling pathways (Tgfβ/Smad, mTor and Fgf/Mapk), calcium handling, proliferation regulators and modifiers of LINC complex function in a cardiac specific Lmna DCM model. Positive candidates for improved cardiac function were further assessed by survival analysis. Suppressive roles and mechanisms of these candidates in ameliorating Lmna DCM were dissected by comparing marker gene expression, Tgfβ signaling pathway activation, fibrosis, inflammation, proliferation and DNA damage. Furthermore, transcriptome profiling compared the differences between Lamin A and Lamin C treatment. Results Cardiac function was restored by several positive candidates (Smad3, Yy1, Bmp7, Ctgf, aYAP1, Sun1, Lamin A, and Lamin C), which significantly correlated with suppression of HF/fibrosis marker expression and cardiac fibrosis in Lmna DCM. Lamin C or Sun1 shRNA administration achieved consistent, prolonged survival which highly correlated with reduced heart inflammation and DNA damage. Importantly, Lamin A treatment improved but could not reproduce long term survival, and Lamin A administration to healthy hearts itself induced DCM. Mechanistically, we identified this lapse as caused by a dose-dependent toxicity of Lamin A, which was independent from its maturation. Conclusions In vivo candidate evaluation revealed that supplementation of Lamin C or knockdown of Sun1 significantly suppressed Lmna DCM and achieve prolonged survival. Conversely, Lamin A supplementation did not rescue long term survival and may impart detrimental cardiotoxicity risk. This study highlights a potential of advancing Lamin C and Sun1 as therapeutic targets for the treatment of LMNA DCM. Graphical Abstract
Article
X‐linked nephrogenic diabetes insipidus (X‐NDI) is a rare congenital disease caused by inactivating mutations of the vasopressin type‐2 receptor (AVPR2), characterized by impaired renal concentrating ability, dramatic polyuria, polydipsia and risk of dehydration. The disease, which still lacks a cure, could benefit from the pharmacologic stimulation of other GPCRs, activating the cAMP‐intracellular pathway in the kidney cells expressing the AVPR2. On the basis of our previous studies, we here hypothesized that the β3‐adrenergic receptor could be such an ideal candidate. We evaluated the effect of continuous 24 h stimulation of the β3‐AR with the agonist BRL37344 and assessed the effects on urine output, urine osmolarity, water intake and the abundance and activation of the key renal water and electrolyte transporters, in the mouse model of X‐NDI. Here we demonstrate that the β3‐AR agonism exhibits a potent antidiuretic effect. The strong improvement in symptoms of X‐NDI produced by a single i.p. injection of BRL37344 (1 mg/kg) was limited to 3 h but repeated administrations in the 24 h, mimicking the effect of a slow‐release preparation, promoted a sustained antidiuretic effect, reducing the 24 h urine output by 27%, increasing urine osmolarity by 25% and reducing the water intake by 20%. At the molecular level, we show that BRL37344 acted by increasing the phosphorylation of NKCC2, NCC and AQP2 in the renal cell membrane, thereby increasing electrolytes and water reabsorption in the kidney tubule of X‐NDI mice. Taken together, these data suggest that human β3‐AR agonists might represent an effective possible treatment strategy for X‐NDI.
Article
Background Information Lamins are type V intermediate filament proteins underlying the inner nuclear membrane which provide structural rigidity to the nucleus, tether the chromosomes, maintain nuclear homeostasis, and remain dynamically associated with developmentally regulated regions of the genome. A large number of mutations particularly in the LMNA gene encoding lamin A/C results in a wide array of human diseases, collectively termed as laminopathies. Dilated Cardiomyopathy (DCM) is one such laminopathic cardiovascular disease which is associated with systolic dysfunction of left or both ventricles leading to cardiac arrhythmia which ultimately culminates into myocardial infarction. Results In this work, we have unraveled the epigenetic landscape to address the regulation of gene expression in mouse myoblast cell line in the context of the missense mutation LMNA 289A<G (Lys97Glu) that is found in DCM‐afflicted patient with severe symptoms. Significant changes in H3‐specific epigenetic modifications indicated a dysregulation in transcription machinery which was investigated by RNA sequencing analysis. The major pathways involved in IL‐17 signaling, cellular response to interferon‐beta and gamma, cytokine production, and related pathways are found to be downregulated. Analysis of the promoter sequences of the genes in the abovementioned pathways led us to the master regulator NF‐κB and its regulatory network. Conclusions We report here for the first time that there is a significant downregulation of the NF‐κB pathway, which has been implicated in cardio‐protection elsewhere. Significance This provides a new pathophysiological explanation that correlates an LMNA mutation and dilated cardiomyopathy.
Article
Background To date, no studies have investigated the association between red blood cell distribution width (RDW)-to-platelet ratio (RPR) and readmission rates among patients with heart failure (HF). As such, the present study aimed to examine the relationship between RPR and readmission rates in patients with HF. Methods Data for this study were obtained from the Fourth People's Hospital (Zigong, Sichuan Province, China). Patients were diagnosed with HF in accordance with European Society of Cardiology criteria. The primary outcome was the 28-day readmission rate. Various logistic regression models were constructed to explore the association between RPR and the 28-day readmission rate. Results The study comprised 1978 patients with HF, with a 28-day readmission rate of 6.98%. RPR emerged as an independent risk factor for 28-day readmission, evidenced by consistent results across the various regression-adjusted models. The covariate-adjusted propensity score model demonstrated that every 0.1 increase in RPR was associated with an 8.2% increase in 28-day readmission rate (odds ratio [OR] 1.082 [95% confidence interval (CI) 1.012–1.158]; P = 0.0212). Similarly, each 0.1 change in RPR was associated with a 9.8% (OR 1.098 [95% CI 1.014–1.188]) and 7.3% (OR 1.073 [95% CI 0.991–1.161]) increase in 3- and 6-month readmission rates, respectively. However, RPR was not statistically associated with the 6-month readmission rate. Curve fit plots illustrated a nonlinear positive correlation between RPR and 28-day, and 3- and 6-month readmissions. Moreover, the effects of RPR on 28-day, and 3- and 6-month readmission rates remained robust across subgroup variables in stratified analysis. Finally, the effect sizes of pooled multiply imputed data were consistent with the original data, suggesting robust results. Conclusion RPR was an independent risk factor for 28-day readmission among patients with HF and also demonstrated modest predictive value for readmissions at 3 and 6 months, despite being non-significant for the 6-month readmission rate. Early identification of patients with HF with elevated RPR would facilitate management and may confer favorable effects on prognosis.
Article
Mandibuloacral dysplasia type A (MADA) is a rare genetic progeroid syndrome associated with lamin A/C (LMNA) mutations. Pathogenic mutations of LMNA result in nuclear structural abnormalities, mesenchymal tissue damage, and progeria phenotypes. However, it remains elusive how LMNA mutations cause mesenchymal-derived cell senescence and disease development. Here, we established an in vitro senescence model using induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) from MADA patients with homozygous LMNA p.R527C mutation. When expanded to passage 13 in vitro, R527C iMSCs exhibited marked senescence and attenuation of stemness potential, accompanied by immunophenotypic changes. Transcriptome and proteome analysis revealed that cell cycle, DNA replication, cell adhesion, and inflammation might play important roles in senescence. In-depth evaluation of changes in extracellular vesicle (EV) derived iMSCs during senescence revealed that R527C iMSC-EVs could promote surrounding cell senescence by carrying pro-senescence microRNAs (miRNAs), including a novel miRNA called miR-311, which can serve as a new indicator for detecting chronic and acute MSC senescence and play a role in promoting senescence. Overall, this study advanced our understanding of the impact of LMNA mutations on MSC senescence and provided novel insights into MADA therapy as well as the link between chronic inflammation and aging development.
Article
Full-text available
Hutchinson–Gilford progeria syndrome (HGPS) causes premature aging in children, with adipose tissue, skin and bone deterioration, and cardiovascular impairment. In HGPS cells and mouse models, high levels of interleukin‐6, an inflammatory cytokine linked to aging processes, have been detected. Here, we show that inhibition of interleukin‐6 activity by tocilizumab, a neutralizing antibody raised against interleukin‐6 receptors, counteracts progeroid features in both HGPS fibroblasts and LmnaG609G/G609G progeroid mice. Tocilizumab treatment limits the accumulation of progerin, the toxic protein produced in HGPS cells, rescues nuclear envelope and chromatin abnormalities, and attenuates the hyperactivated DNA damage response. In vivo administration of tocilizumab reduces aortic lesions and adipose tissue dystrophy, delays the onset of lipodystrophy and kyphosis, avoids motor impairment, and preserves a good quality of life in progeroid mice. This work identifies tocilizumab as a valuable tool in HGPS therapy and, speculatively, in the treatment of a variety of aging‐related disorders.
Article
Full-text available
Heart failure exhibits remarkable pathophysiologic heterogeneity. A large body of evidence suggests that regardless of the underlying etiology, heart failure is associated with induction of cytokines and chemokines that may contribute to the pathogenesis of adverse remodeling, and systolic and diastolic dysfunction. The pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6 have been extensively implicated in the pathogenesis of heart failure. Inflammatory cytokines modulate phenotype and function of all myocardial cells, suppressing contractile function in cardiomyocytes, inducing inflammatory activation in macrophages, stimulating microvascular inflammation and dysfunction, and promoting a matrix-degrading phenotype in fibroblasts. Moreover, cytokine-induced growth factor synthesis may exert chronic fibrogenic actions contributing to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). In addition to their role in adverse cardiac remodeling, some inflammatory cytokines may also exert protective actions on cardiomyocytes under conditions of stress. Chemokines, such as CCL2, are also upregulated in failing hearts and may stimulate recruitment of pro-inflammatory leukocytes, promoting myocardial injury, fibrotic remodeling, and dysfunction. Although experimental evidence suggests that cytokine and chemokine targeting may hold therapeutic promise in heart failure, clinical translation remains challenging. This review manuscript summarizes our knowledge on the role of TNF-α, IL-1, IL-6, and CCL2 in the pathogenesis of heart failure, and discusses the promises and challenges of targeted anti-cytokine therapy. Dissection of protective and maladaptive cellular actions of cytokines in the failing heart, and identification of patient subsets with overactive or dysregulated myocardial inflammatory responses are required for design of successful therapeutic approaches.
Article
Full-text available
Mutations of Lamin A/C gene (LMNA) cause laminopathies, a group of disorders associated with a wide spectrum of clinically distinct phenotypes, affecting different tissues and organs. Heart involvement is frequent and leads to cardiolaminopathy LMNA-dependent cardiomyopathy (LMNA-CMP), a form of dilated cardiomyopathy (DCM) typically associated with conduction disorders and arrhythmias, that can manifest either as an isolated event or as part of a multisystem phenotype. Despite the recent clinical and molecular developments in the field, there is still lack of knowledge linking specific LMNA gene mutations to the distinct clinical manifestations. Indeed, the severity and progression of the disease have marked interindividual variability, even amongst members of the same family. Studies conducted so far have described Lamin A/C proteins involved in diverse biological processes, that span from a structural role in the nucleus to the regulation of response to mechanical stress and gene expression, proposing various mechanistic hypotheses. However, none of those is per se able to fully justify functional and clinical phenotypes of LMNA-CMP; therefore, the role of Lamin A/C in cardiac pathophysiology still represents an open question. In this review we provide an update on the state-of-the-art studies on cardiolaminopathy, in the attempt to draw a line connecting molecular mechanisms to clinical manifestations. While investigators in this field still wonder about a clear genotype/phenotype correlation in LMNA-CMP, our intent here is to recapitulate common mechanistic hypotheses that link different mutations to similar clinical presentations.
Article
Full-text available
Laminopathies are a wide and heterogeneous group of rare human diseases caused by mutations of the LMNA gene or related nuclear envelope genes. The variety of clinical phenotypes and the wide spectrum of histopathological changes among patients carrying an identical mutation in the LMNA gene make the prognostic process rather difficult, and classical genetic screens appear to have limited predictive value for disease development. The aim of this study was to evaluate whether a comprehensive profile of circulating cytokines may be a useful tool to differentiate and stratify disease subgroups, support clinical follow-ups and contribute to new therapeutic approaches. Serum levels of 51 pro- and anti-inflammatory molecules, including cytokines, chemokines and growth factors, were quantified by a Luminex multiple immune-assay in 53 patients with muscular laminopathy (Musc-LMNA), 10 with non-muscular laminopathy, 22 with other muscular disorders and in 35 healthy controls. Interleukin-17 (IL-17), granulocyte colony-stimulating factor (G-CSF) and transforming growth factor beta (TGF-β2) levels significantly discriminated Musc-LMNA from controls; interleukin-1β (IL-1β), interleukin-4 (IL-4) and interleukin-8 (IL-8) were differentially expressed in Musc-LMNA patients compared to those with non-muscular laminopathies, whereas IL-17 was significantly higher in Musc-LMNA patients with muscular and cardiac involvement. These findings support the hypothesis of a key role of the immune system in Musc-LMNA and emphasize the potential use of cytokines as biomarkers for these disorders.
Article
Full-text available
Cardiomyopathy caused by A-type lamins gene (LMNA) mutations (LMNA cardiomyopathy) is associated with dysfunction of the heart, often leading to heart failure. LMNA cardiomyopathy is highly penetrant with bad prognosis with no specific therapy available. Searching for alternative ways to halt the progression of LMNA cardiomyopathy, we studied the role of calcium homeostasis in the evolution of this disease. We showed that sarcolipin, an inhibitor of the sarco/endoplasmic reticulum (SR) Ca²⁺ ATPase (SERCA) was abnormally elevated in the ventricular cardiomyocytes of mutated mice compared with wild type mice, leading to an alteration of calcium handling. This occurs early in the progression of the disease, when the left ventricular function was not altered. We further demonstrated that down regulation of sarcolipin using adeno-associated virus (AAV) 9-mediated RNA interference delays cardiac dysfunction in mouse model of LMNA cardiomyopathy. These results showed a novel role for sarcolipin on calcium homeostasis in heart and open perspectives for future therapeutic interventions to LMNA cardiomyopathy.
Article
Full-text available
Background Heart failure (HF) is one of the most significant causes of morbidity and mortality for the cardiovascular risk population. We found previously that extracellular HSP70 (heat shock protein) is an important trigger in cardiac hypertrophy and fibrosis, which are associated with the development of heart dysfunction. However, the potential role of HSP70 in response to HF and whether it could be a target for the therapy of HF remain unknown. Methods and Results An HF mouse model was generated by a single IP injection of doxorubicin at a dose of 15 mg/kg. Ten days later, these mice were treated with an HSP70 neutralizing antibody for 5 times. We observed that doxorubicin treatment increased circulating HSP70 and expression of HSP70 in myocardium and promoted its extracellular release in the heart. Blocking extracellular HSP70 activity by its antibody significantly ameliorated doxorubicin‐induced left ventricular dilation and dysfunction, which was accompanied by a significant inhibition of cardiac fibrosis. The cardioprotective effect of the anti‐HSP70 antibody was largely attributed to its ability to promote the resolution of myocardial inflammation, as evidenced by its suppression of the toll‐like receptor 2–associated signaling cascade and modulation of the intracellular distribution of the p50 and p65 subunits of nuclear factor‐κB. Conclusions Extracellular HSP70 serves as a noninfectious inflammatory factor in the development of HF, and blocking extracellular HSP70 activity may provide potential therapeutic benefits for the treatment of HF.
Article
Full-text available
Background Systemic inflammation is a strong predictor of atrial fibrillation. A key role for electrical remodeling is increasingly recognized, and experimental data suggest that inflammatory cytokines can directly affect connexins resulting in gap‐junction dysfunction. We hypothesized that systemic inflammation, regardless of its origin, promotes atrial electric remodeling in vivo, as a result of cytokine‐mediated changes in connexin expression. Methods and Results Fifty‐four patients with different inflammatory diseases and elevated C‐reactive protein were prospectively enrolled, and electrocardiographic P‐wave dispersion indices, cytokine levels (interleukin‐6, tumor necrosis factor‐α, interleukin‐1, interleukin‐10), and connexin expression (connexin 40, connexin 43) were measured during active disease and after reducing C‐reactive protein by >75%. Moreover, peripheral blood mononuclear cells and atrial tissue specimens from an additional sample of 12 patients undergoing cardiac surgery were evaluated for atrial and circulating mRNA levels of connexins. Finally, in vitro effects of interleukin‐6 on connexin expression were studied in HL‐1 mouse atrial myocytes. In patients with active inflammatory diseases, P‐wave dispersion indices were increased but rapidly decreased within days when C‐reactive protein normalizes and interleukin‐6 levels decline. In inflammatory disease patients, both P‐wave dispersion indices and interleukin‐6 changes were inversely associated with circulating connexin levels, and a positive correlation between connexin expression in peripheral blood mononuclear cells and atrial tissue was demonstrated. Moreover, interleukin‐6 significantly reduced connexin expression in HL‐1 cells. Conclusions Our data suggest that regardless of specific etiology and organ localization, systemic inflammation, via interleukin‐6 elevation, rapidly induces atrial electrical remodeling by down‐regulating cardiac connexins. Although transient, these changes may significantly increase the risk for atrial fibrillation and related complications during active inflammatory processes.
Article
Full-text available
Mutations in LMNA, which encodes the nuclear proteins Lamin A/C, can cause cardiomyopathy and conduction disorders. Here, we employ induced pluripotent stem cells (iPSCs) generated from human cells carrying heterozygous K219T mutation on LMNA to develop a disease model. Cardiomyocytes differentiated from these iPSCs, and which thus carry K219T-LMNA, have altered action potential, reduced peak sodium current and diminished conduction velocity. Moreover, they have significantly downregulated Nav1.5 channel expression and increased binding of Lamin A/C to the promoter of SCN5A, the channel’s gene. Coherently, binding of the Polycomb Repressive Complex 2 (PRC2) protein SUZ12 and deposition of the repressive histone mark H3K27me3 are increased at SCN5A. CRISPR/Cas9-mediated correction of the mutation re-establishes sodium current density and SCN5A expression. Thus, K219T-LMNA cooperates with PRC2 in downregulating SCN5A, leading to decreased sodium current density and slower conduction velocity. This mechanism may underlie the conduction abnormalities associated with LMNA-cardiomyopathy.
Article
Mutations in the lamin A/C gene (LMNA), which encodes A-type lamins, cause several diseases called laminopathies, the most common of which is dilated cardiomyopathy with muscular dystrophy. The role of Ca2+ regulation in these diseases remain poorly understood. We now show biochemical remodeling of the ryanodine receptor (RyR)/intracellular Ca2+ release channel in heart samples from human subjects with LMNA mutations, including protein kinase A-catalyzed phosphorylation, oxidation and depletion of the stabilizing subunit calstabin. In the LmnaH222P/H222P murine model of Emery-Dreifuss muscular dystrophy caused by LMNA mutation, we demonstrate an age-dependent biochemical remodeling of RyR2 in heart and RyR1 in skeletal muscle. This RyR remodeling is associated with heart and skeletal muscle dysfunction. Defective heart and muscle function are ameliorated by treatment with a novel Rycal small molecule drug (S107) that fixes ‘leaky’ RyRs. SMAD3 phosphorylation is increased in hearts and diaphragms of LmnaH222P/H222P mice, which enhances NADPH oxidase binding to RyR channels, contributing to their oxidation. There is also increased generalized protein oxidation, increased calcium/calmodulin-dependent protein kinase II-catalyzed phosphorylation of RyRs and increased protein kinase A activity in these tissues. Our data show that RyR remodeling plays a role in cardiomyopathy and skeletal muscle dysfunction caused by LMNA mutation and identify these Ca2+ channels as a potential therapeutic target.
Article
Premature ventricular complexes (PVCs) are extremely common, found in the majority of individuals undergoing long-term ambulatory monitoring. Increasing age, a taller height, a higher blood pressure, a history of heart disease, performance of less physical activity, and smoking each predict a greater PVC frequency. Although the fundamental causes of PVCs remain largely unknown, potential mechanisms for any given PVC include triggered activity, automaticity, and reentry. PVCs are commonly asymptomatic but can also result in palpitations, dyspnea, presyncope, and fatigue. The history, physical examination, and 12-lead ECG are each critical to the diagnosis and evaluation of a PVC. An echocardiogram is indicated in the presence of symptoms or particularly frequent PVCs, and cardiac magnetic resonance imaging is helpful when the evaluation suggests the presence of associated structural heart disease. Ambulatory monitoring is required to assess PVC frequency. The prognosis of those with PVCs is variable, with ongoing uncertainty regarding the most informative predictors of adverse outcomes. An increased PVC frequency may be a risk factor for heart failure and death, and the resolution of systolic dysfunction after successful catheter ablation of PVCs demonstrates that a causal relationship can be present. Patients with no or mild symptoms, a low PVC burden, and normal ventricular function may be best served with simple reassurance. Either medical treatment or catheter ablation are considered first-line therapies in most patients with PVCs associated with symptoms or a reduced left ventricular ejection fraction, and patient preference plays a role in determining which to try first. If medical treatment is selected, either β-blockers or nondihydropyridine calcium channel blockers are reasonable drugs in patients with normal ventricular systolic function. Other antiarrhythmic drugs should be considered if those initial drugs fail and ablation has been declined, has been unsuccessful, or has been deemed inappropriate. Catheter ablation is the most efficacious approach to eradicate PVCs but may confer increased upfront risks. Original research remains necessary to identify individuals at risk for PVC-induced cardiomyopathy and to identify preventative and therapeutic approaches targeting the root causes of PVCs to maximize effectiveness while minimizing risk.