IMR Press / RCM / Volume 23 / Issue 3 / DOI: 10.31083/j.rcm2303108
Open Access Review
Restrictive cardiomyopathy: from genetics and clinical overview to animal modeling
Show Less
1 College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
2 Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
3 Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
4 Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
*Correspondence: mchintanaphol@gmail.com (Michelle Chintanaphol); epurevja@uthsc.edu (Enkhsaikhan Purevjav)
Academic Editors: Sophie Mavrogeni and Robert C Hendel
Rev. Cardiovasc. Med. 2022, 23(3), 108; https://doi.org/10.31083/j.rcm2303108
Submitted: 18 August 2021 | Revised: 8 October 2021 | Accepted: 25 October 2021 | Published: 17 March 2022
(This article belongs to the Special Issue State-of-the-Art Cardiovascular Medicine in the USA 2022)
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Restrictive cardiomyopathy (RCM), a potentially devastating heart muscle disorder, is characterized by diastolic dysfunction due to abnormal muscle relaxation and myocardial stiffness resulting in restrictive filling of the ventricles. Diastolic dysfunction is often accompanied by left atrial or bi-atrial enlargement and normal ventricular size and systolic function. RCM is the rarest form of cardiomyopathy, accounting for 2–5% of pediatric cardiomyopathy cases, however, survival rates have been reported to be 82%, 80%, and 68% at 1-, 2-, and 5-years after diagnosis, respectively. RCM can be idiopathic, familial, or secondary to a systemic disorder, such as amyloidosis, sarcoidosis, and hereditary hemochromatosis. Approximately 30% of cases are familial RCM, and the genes that have been linked to RCM are cTnT, cTnI, MyBP-C, MYH7, MYL2, MYL3, DES, MYPN, TTN, BAG3, DCBLD2, LNMA, and FLNC. Increased Ca2+ sensitivity, sarcomere disruption, and protein aggregates are some of the few mechanisms of pathogenesis that have been revealed by studies utilizing cell lines and animal models. Additional exploration into the pathogenesis of RCM is necessary to create novel therapeutic strategies to reverse restrictive cardiomyopathic phenotypes.

Keywords
cardiomyopathy
restrictive cardiomyopathy
restrictive physiology
mutation
1. Introduction

Restrictive cardiomyopathy (RCM) is a rare form of heart muscle disease, characterized by diastolic dysfunction provoked from myocardial stiffness in the setting of normal or reduced biventricular diastolic and systolic volumes [1, 2]. Impaired diastolic function leads to left atrial or biatrial enlargement and elevated atrial pressures. RCM may eventually affect ventricular chamber size and systolic function. These patients can progress to heart failure with preserved ejection fraction (HFpEF) or heart failure with reduced ejection fraction if left untreated [1, 3]. RCM accounts for 2–5% of all diagnosed cardiomyopathies; however, it has the worst outcome among cardiomyopathies with up to 50% of deaths occurring within 2 years after diagnosis in children [4]. With a median survival of 2.2 years without heart transplantation, RCM remains a disease that is largely unexplored and with limited treatment options [5]. Advancements in molecular genetics and improved assessment by radiography and echocardiography throughout the past few decades have expanded diagnostic capabilities and understanding of pathophysiology underlying RCM.

The term cardiomyopathy was first defined by Brigden in 1957 to describe uncommon isolated and idiopathic non-coronary diseases of the myocardium with familial patterns in some families that were studied [6]. The cases of pathological asymmetric hypertrophy were first described as an obstructive cardiomyopathy by Teare in 1958 [7], and familial cases of this condition with notable arrhythmia and sudden death have subsequently been published [8]. Later, three principal clinical presentations have been used to classify cardiomyopathies: (1) congestive heart failure and cardiac enlargement with dilated and flabby myocardium, (2) cardiac constriction with stiff inelastic myocardium and associated endocardial and pericardial involvement named as constrictive cardiomyopathy, and (3) cardiac obstruction with specific asymmetrical hypertrophy of the septum (obstructive cardiomyopathy) [9]. Goodwin and Oakley distinguished the obliterative (constrictive) subtype of cardiomyopathy as a rare heart muscle disease specific to amyloid and leukemic infiltration of polyarthritis nodosa [10]. As a clinically recognizable disorder, the term “primary RCM” was introduced in 1977 by Chew et al. [11]. The authors described 11 RCM patients with endomyocardial fibrosis, who shared a common diastolic abnormality with restriction of ventricular filling and stroke output as a main feature that was not seen in other primary cardiomyopathies nor in amyloid heart disease. Along with dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), RCM was first classified as a cardiomyopathy in 1980 by the World Health Organization (WHO) and the International Society and Federation of Cardiology (ISFC) [12]. This was the first classification of cardiac heart muscle diseases of unknown cause based on altered hemodynamics (restrictive ventricular filling) rather than pathomorphological characteristics. Later in 1996, the WHO and the Federation of Cardiology Task Force added arrhythmogenic right ventricular cardiomyopathy (ARVC) to the list of cardiomyopathies [13]. With the advent of molecular genetics and improvements in diagnostic modalities, the perspective on cardiomyopathies changed to incorporate causative genetic mutations and modifier genes to explain pathologic mechanisms. Multiple genetic mutations have been found in patients with RCM and occur primarily in genes encoding sarcomeric and cytoskeletal proteins, which are influenced by mechanosensing and mechanotransducing proteins [14]. In the time since 1996, the American Heart Association (AHA) in 2006 and the European Society of Cardiology (ESC) in 2008 updated their statements regarding the classification of cardiomyopathies [2, 15].

The difficulty in classifying cardiomyopathies lies with the overlap in morphological and physiological abnormalities, genetic mutations, and the evolving nature of the disease. The AHA consensus panel gives a broad definition of cardiomyopathies and categorizes cardiomyopathies as primary or secondary, depending on whether the disease is restricted to heart tissue or whether cardiac dysfunction results from a systemic disorder. Under the AHA classification system, primary cardiomyopathies are mostly of idiopathic or genetic origin, whereas secondary cardiomyopathies have an underlying cause. Secondary cardiomyopathies include amyloidosis, sarcoidosis, storage disorders, toxicity from drugs or chemical agents, endomyocardial fibrosis, endocrine disorders, and more. In this classification scheme, the last of the cardiomyopathies to be identified in 2006 was left ventricular noncompaction (LVNC). Under the AHA definition, RCM is based on hemodynamic dysfunction of restrictive physiology and categorized as a primary restrictive nonhypertrophied cardiomyopathy of genetic or nongenetic etiologies, whereas HCM and DCM depend on morphological abnormalities [15].

On the other hand, the ESC definition is based on both morphological and functional abnormalities that are important for diagnosis and not caused by myocardial ischemia from coronary artery disease, hypertension, valvular disease, or congenital heart disease, with no focus on the primary or secondary cause. Each classified cardiomyopathy is divided into subsets of familial or genetic cardiomyopathies and non-familial or non-genetic cardiomyopathies. Cardiomyopathies are considered familial if more than one family member presents with the same pathological phenotype caused by a pathogenic genetic variant. Both familial and non-familial cardiomyopathies are further subdivided into categories, one of which is “disease sub-type”, in which myocardial dysfunction is due to a systemic disorder, similar to the AHA secondary cardiomyopathy classification [2].

Etiologically, RCM can be idiopathic, familial (primarily autosomal dominant inheritance, but also autosomal recessive or X-linked), or secondary to a systemic disorder [16]. Secondary diseases can be characterized as infiltrative or non-infiltrative cardiomyopathies, storage disorders, fibrotic injury, or endomyocardial damage [16]. Infiltrative RCMs include amyloidosis, sarcoidosis, Gaucher’s disease, and Hurler syndrome while non-infiltrative cardiomyopathies include idiopathic RCM and scleroderma. Storage diseases that may present with RCM are Fabry’s disease, glycogen storage disorders, and hemochromatosis. The endocardium can be damaged due to endomyocardial fibrosis, hyper-eosinophilic syndromes, metastatic tumors, radiation, sickle cell disease, or drugs such as anthracyclines and other chemotherapeutic agents, ergotamine, and chloroquine [17]. In adults less than 30 years of age, RCM is most commonly linked to a wide variety of systemic disorders while the associated disorders for adults with RCM over 65 years of age are limited to amyloidosis, hemochromatosis, and radiation-induced endomyocardial damage [17].

This article will describe the epidemiology and clinical presentation of RCM, as well as the genetic basis and pathophysiology of the disease. We will also highlight family studies and animal models that have been crucial in providing insights into the pathological and pathophysiological mechanisms of RCM.

2. Epidemiology

The incidence and prevalence of RCM remains unclear and variable due to differences in geographic region, sex, and ethnicity parameters. Due to its rarity and overlap in HCM, DCM, and LVNC phenotypes, a lack of epidemiological studies in RCM persists. Based on a study conducted between 1982 and 1997, RCM is the least common cardiomyopathy and accounts for approximately 2–5% of all cardiomyopathies in United States [18]. In pediatric cases, RCM accounted for 4.5% of cardiomyopathy cases (152 of 3375 children under 18 years of age) as reported by the Pediatric Cardiomyopathy Registry (PCMR) database, inclusive of 98 institutions in the United States and Canada between 1990 and 2008. Out of 152 pediatric RCM cases, 101 cases were solely RCM diagnoses as opposed to 51 cases which were a mixed RCM and HCM phenotype. Ninety-three percent of the purely RCM cases diagnosed in childhood were idiopathic in nature, 14% of the RCM patients had a family history of cardiomyopathy, 1% of cases were related to inborn errors of metabolism, and no RCM cases were secondary to a neuromuscular disorder. The mean age of pediatric diagnoses was 6.2 years [4]. A 10-year report under the National Australian Childhood Cardiomyopathy reported that 2.5% of pediatric cardiomyopathy cases between 1987 and 1996 were RCM diagnoses. Half of those patients presented with congestive heart failure (CHF). Unlike pediatric cases of DCM or HCM, which mostly present at infancy, the median age of presentation for pediatric RCM in this cohort was 3 years of age [19]. In New Delhi, India, the epidemiology of cardiomyopathy study (EPOCH) at the All India Institute of Medical Sciences reported 30 out of 190 cardiomyopathy cases (1.6%) were RCM and 63% of those RCM patients were male [20]. However, the true incidence and prevalence of RCM is unknown, as echocardiography is not a common assessment in the general population worldwide.

It is less difficult to track epidemiology for disorders underlying secondary RCM, although these systemic disorders do not equate to patients having idiopathic RCM. The incidence of cardiac amyloidosis is 3–9 cases per million people and is characterized by the extracellular deposition of amyloid fibrils in the heart. According to the most recent position statement of the ESC on cardiac amyloidosis, more than 98% of currently diagnosed cardiac amyloidosis results from fibrils composed of monoclonal immunoglobulin light chains (AL) or transthyretin (ATTR), either in its hereditary or acquired form [21]. Around 60–80% of patients with AL amyloidosis have cardiac involvement [22, 23]. AL amyloidosis is more common in male patients and at ages 55 to 60 years and death in 75% of the 393 patients was attributed to cardiac amyloidosis, including sudden death in 25% [24]. In contrast, ATTR amyloidosis is more common in patients older than 80 years [3, 25] and is prevalent among patients with heart failure, especially HFpEF [26]. However, the true prevalence of RCM in ATTR remains largely unknown [27].

Sarcoidosis, a multi-organ disease associated with secondary RCM, is characterized by inflammation and noncaseating granulomas [28]. Incidence and prevalence of sarcoidosis is high in Sweden, Norway, Denmark, Japan, and the United States [29, 30]. Scandinavian countries report an incidence of 50 to 60 cases per 100,000 per year [31]. The incidence rate of sarcoidosis was highest in black women in the United States: 46 per 100,000 per year according to the Nurses’ Health Study II and 17.8 per 100,000 per year based on the Optum health insurance medical claims database [32, 33]. Although clinical signs of cardiac involvement are present in 5% of patients with sarcoidosis, autopsy studies have demonstrated the presence of cardiac granuloma and granulomatous inflammation in up to 25% of autopsy specimens [34].

Storage diseases such as hemochromatosis (iron storage disorder), Fabry disease (lysosomal storage disorder) and Danon and Pompe diseases (glycogen storage disorders) are genetic disorders associated with diastolic dysfunction and RCM or HCM [35]. It is difficult to accurately estimate the incidence and prevalence of RCM associated with those genetic disorders [23].

3. Overview of clinical manifestations

Overall, the clinical presentation of RCM is nonspecific. Initially, patients are asymptomatic and do not seek medical attention unless they have a family history of cardiomyopathy or symptoms of fatigue, dyspnea, or exercise intolerance [36]. In both pediatric and adult patients with RCM, dyspnea is the most common symptom [37]. Pediatric patients with RCM usually present with fatigue, syncope, hepatomegaly, cardiomegaly on radiograph, and failure to thrive. Initial presentation of RCM in pediatric patients are typically pulmonary related, with frequent lung infections and reactive airway disease [38]. Arrhythmias or atrioventricular block increase risk for sudden cardiac death, and syncope may anticipate ischemic complications, arrhythmias, thromboembolism, and sudden death. Rivenes et al. [39] reported on 18 childhood RCM cases and identified that presentation with syncope and/or signs of ischemia had high sudden death rates of 28% with an annual mortality rate of 7% in the cohorts studied. Routine screening for ischemia in all pediatric patients with RCM should include a baseline ECG and Holter monitoring with digital ST-segment analysis as screening tools (Fig. 1, Ref. [39]).

Fig. 1.

ECG and Holter monitor tracing in pediatric patient with RCM and chest pain who sustained an acute myocardial infarction. (A) ECG tracing, demonstrating bi-atrial enlargement and ST-segment depression in inferior lateral and lateral precordial leads (arrows, evidence of ischemia). (B) Sinus tachycardia and ST-segment depression precede degeneration to torsade de pointes. Arrow indicates inciting “R-on-T” phenomenon. (C) Tracing obtained several seconds later, depicting classic torsade de pointes pattern [39].

Adult patients more commonly present with fatigue, dyspnea, edema, and limited exercise tolerance [16, 40, 41]. In a study cohort (1979–1996) of 94 patients with idiopathic RCM, the most common symptoms were dyspnea (71%), history of edema (46%), palpitations (33%), fatigue (32%), orthopnea (22%), and chest pain (22%). Typical physical findings were jugular venous distension (52%), systolic murmur (49%), S3 gallop (27%), and pulmonary rales (18%). Fifty-three percent of patients were categorized as NYHA class II [42]. In advanced stages of the disease, patients may develop thromboembolism, arrhythmias, heart failure, and cardiac cirrhosis [25]. RCM with HFpEF most commonly presents with heart failure symptoms such as distended jugular veins, pulsus paradoxus, pleural effusion, bilateral edema, respiratory rales, hepatomegaly, ascites, and an S3 and S4 gallop [40].

Conduction defects can be explained by the restrictive physiology of RCM, which is characterized by increased ventricular pressure caused by myocardial stiffness and impaired ventricular filling. In contrast to restrictive filling seen in other pathologies, diastolic dysfunction with abnormal relaxation in RCM is accompanied by near normal diastolic and systolic volumes, contractile function, and wall thickness [2]. Wall thickness is increased in infiltrative diseases and additional phenotypes such as HCM. In advanced stages of RCM, systolic function decreases [4]. The abnormal ventricular relaxation leads to a buildup of blood from impaired filling and atrial enlargement (Fig. 2). Bi-atrial enlargement and left ventricular stiffness may cause arrhythmias and thromboembolism from stasis of flow and resistance in pulmonary venous flow from increased atrial pressures and symptoms of heart failure. Mitral and tricuspid regurgitation can develop and consequently exacerbate atrial enlargement [17]. Fibrosis, myocardial stiffness, and the resultant restrictive remodeling are responsible for poor prognosis, but the basis of this remodeling remains to be fully understood.

Fig. 2.

Echocardiographic and ECG features of RCM. (A) Image of apical two-dimensional echocardiography in 4-chamber view. Severe right (RA) and left (LA) atrial enlargement relative to normal biventricular (RV and LV) size and thickness is captured. (B) ECG tracing, demonstrating bi-atrial enlargement (oval) and nonspecific ST-T wave changes.

4. Diagnosis

Elevated ventricular filling pressures cause left and/or right atrial dilatation and symptoms of heart failure as discussed earlier. RCM must be differentiated from constrictive pericarditis (CP), HCM, and hypertensive heart disease. The differential diagnosis is established by physical examination, clinical laboratory testing, electrocardiogram, echocardiography, and cardiac magnetic resonance (CMR) with late gadolinium enhancement (LGE) [17, 40]. Myocardial stiffness eventually increases systemic filling pressures and decreases stroke volume, which in turn may affect kidney function [17]. To evaluate for heart failure and the degree of which other organs are affected, clinical labs can be sent for hematocrit, comprehensive metabolic panel, creatinine, blood urea nitrogen, urine protein, liver function panel, brain natriuretic peptide (BNP) or N-terminal pro-BNP (Nt-proBNP), and troponin T or I [40]. BNP/Nt-proBNP is markedly elevated in patients with RCM in comparison to patients with CP [43]. In primary systemic amyloidosis, measurements of Nt-proBNP can be used as a prognostic marker for advanced disease and survival rates [44]. Electrocardiogram (ECG) is often employed in the initial assessment of cardiac disorders and a 24-hour Holter monitor test should be obtained in patients with suspected cardiac sarcoidosis to document conduction and rhythm disturbances that may be missed on ECG [23]. Table 1 outlines more specific laboratory testing for systemic diseases associated with secondary RCM [40, 45].

Table 1.Clinical lab testing for systemic disorders associated with restrictive cardiomyopathy.
Disorder Clinical laboratory test
Amyloidosis 24-hour urine monoclonal protein, serum and urine immunofixation with electrophoresis, serum free light chain assay
Cardiac sarcoidosis Angiotensin converting enzyme (ACE)
Hemochromatosis Serum iron, total iron-binding capacity (TIBC), ferritin, liver panel, hemochromatosis gene (HFE) testing
Hyper-eosinophilia Complete blood count (CBC) and differential, peripheral blood smear, anti-neutrophil cytoplasmic antibody (ANCA)

Approximately 99% of ECG readings in RCM patients are abnormal, although only 15–30% of patients present with arrhythmias. Abnormalities are highly variable but must commonly are biphasic P wave in the precordial leads indicative of bi-atrial enlargement, elevated ST segments, Q-wave abnormalities, and notched or biphasic late peaking T waves [37, 46]. Low voltage QRS complexes indicates increased ventricular wall thickness and is a characteristic of AL amyloidosis as well as other infiltrative diseases [25]. Abnormalities in the ST segment and atrioventricular conduction and supraventricular and ventricular arrhythmias may be found in patients with RCM [40, 47]. In approximately 50–70% of patients with amyloidosis, pseudo-infarct patterns are present [48].

According to the 2016 American Society of Echocardiography (ASE) and European Association of Cardiovascular Imaging (EACVI) guidelines, advanced myocardial involvement with grade 3 diastolic dysfunction, associated with poor prognosis, is detected by transmitral spectral Doppler echocardiography (Fig. 3). The restrictive filling pattern is identified by parameters of early diastolic velocity (increased E wave), low or absent late filling velocity (decreased A wave), mitral inflow E/A ratio >2.5, as well as a dip and plateau pattern of early diastolic LV pressure, deceleration time of E velocity <150 msec, isovolumic relaxation time <50 msec, septal and lateral e’ velocity of 3–4 cm/sec with lateral e’ velocity greater than septal e’ velocity, E/e’ ratio >14, and LA volume index >50 mL/m2 [49].

Fig. 3.

M-mode images of mitral valve movement by transthoracic 2-dimensional Doppler echocardiography showing normal diastolic function with E > A wave (A) and a restrictive filling pattern of LV with increased E wave and E/A ratios >2.5 (B).

Diastolic dysfunction increases atrial filling pressures, which consequently impact pulmonary venous flow with a decreased systolic to diastolic pulmonary venous flow ratio [17]. Heart failure with HFpEF is characteristic of RCM, although tissue Doppler can reveal reduced systolic waves, blunted early diastolic waves, and preserved or blunted late diastolic waves [50]. Speckle tracking echocardiography (STE), which can detect myocardial deformities, demonstrates a ratio of LV free wall strain to LV septal strain of 1, reduced longitudinal strain, and normal circumferential strain [49, 51]. Typically, chamber volumes will remain normal and there will be bi-atrial enlargement. Infiltrative types of RCM will have increased LV wall thickness. Pulmonary artery pressure may be increased in very advanced stages of disease [40].

Both RCM and CP have several similarities in echocardiography and clinical symptoms: normal to near normal ventricle size, preserved ejection fraction, dilated atria, increased filling pressure, jugular venous distension, Kussmaul sign, and diastolic sounds [25, 40]. Differentiating between RCM and CP using echocardiography coupled with the advances in cardiac CT and CMR is crucial since CP can be resolved with NSAIDs or pericardiectomy after medical therapy failure [50]. Echocardiography of a patient with RCM will display wall and valvular thickening and “sparkling” myocardium. CP may reveal pericardial thickening and a respiratory ventricular septal shift. While RCM has decreased variation in mitral and tricuspid inflow E velocity, CP has increased variation. In RCM, there may be mitral or tricuspid regurgitation as a consequence of atrial enlargement and restrictive filling [40].

CMR is beneficial in establishing a differential diagnosis among RCM, CP, and infiltrative disorders such as amyloidosis, sarcoidosis, and hemochromatosis. Imaging allows for tissue categorization, including inflammation, and provides high spatial resolution and noninvasive measurement of ventricular volumes. CMR with late gadolinium enhancement (LGE) can assist with diagnosing subtypes of RCM, particularly, amyloidosis and measuring pericardial thickness (Fig. 4). Although not applicable to the RCM diagnosis, CT scans and catheterization with coronary angiography are helpful for eliminating constrictive pericarditis and obstructive coronary artery disease, respectively, from the differential diagnosis. For RCM, chest radiographs show bi-atrial enlargement and in some cases, pulmonary congestion [52].

Fig. 4.

Examples of cardiac magnetic resonance (CMR) in differential diagnosis between restrictive cardiomyopathy (RCM) and amyloidosis. Adopted from Kyriakou et al. 2018 [23]. Cardiac magnetic resonance (CMR) images of patients with cardiac amyloidosis. Amyloid fibril deposition pattern mainly affects subendocardial CMR imaging, leading to a shortened T1 relaxation time and a diffuse LGE of the left ventricular endocardium.

If noninvasive procedures are not decisive, endomyocardial biopsy (EMB) provides histological samples which may be helpful for infiltrative diseases like sarcoidosis [40]. EMB and a positive fat pad aspiration is indicative of cardiac amyloidosis [25]. Cardiac catheterization allows for hemodynamic measurements of filling pressures and pulmonary hypertension. This assessment is helpful in differentiating RCM diagnosis from CP [36]. Right and left ventricular pressure tracing with a dip and plateau pattern (square root sign) and elevated LV and RV diastolic pressures are characteristic of restrictive hemodynamics [16]. A comprehensively reviewed diagnostic algorithm of secondary RCM by Muchtar et al. [3] is illustrated in Fig. 5.

Fig. 5.

Diagnostic algorithm for suspected cardiac amyloidosis. Adopted from Muchtar et al. 2012 [3]. *Alternatively other source tissue other than heart or endomyocardial biopsy (EMB) can be obtained. ᵻThe use of bone scintigraphy can replace EMB.

Genetic testing and counseling are available for patients and their relatives to determine risk of inheritance. Especially, the likelihood and screening of de novo variation(s) should be integrated into genetic counseling of pediatric cardiomyopathy cases [53]. Hereditary forms of cardiac amyloidosis commonly have an adult onset, thus genetic testing of minors is discouraged, although genetic testing is recommended for adult relatives along with genetic counseling [21]. While genetic counseling is more common for patients with HCM, genetic testing should be recommended to families of young patients with a family history of sudden cardiac death, heart failure, and implanted cardioverter defibrillators. In a multinational European study, 43% of patients with RCM were found to have one disease-causing variant [54].

5. Treatment and prognosis

There is no cure for RCM, and patients suffer from poor outcomes. Treatment should center on alleviating heart failure symptoms and treating underlying causes of RCM [23]. Without heart transplantation, survival is limited. Diuretics, with careful monitoring, can relieve volume overload and pulmonary edema or congestion [55]. Since diuretics may worsen cardiac output, patients should restrict dietary sodium [17]. Beta-blockers and calcium channel blockers may be successful in increasing filling time or treating arrhythmias, although should be administered with care since rate-slowing medication may be deleterious. Renin-angiotensin system inhibitors are also not well-tolerated; however, if heart failure advances to systolic dysfunction, angiotensin-receptor blockers and angiotensin II receptor blockers may be employed [3, 25]. Patients with enlarged atria are at increased risk of forming blood clots, and antithrombotic therapy may be administered to patients with high risk for thromboembolisms. Atrial fibrillation (AF) is a common clinical sign in cardiomyopathies, and according to a study by Mizia-Stec et al. [56], prevalence of AF is highest in RCM (52%) compared to rates in DCM, HCM, and ARVC. Stroke incidence is higher in patients with AF versus patients presenting without AF, and if not contraindicated, anticoagulants should be recommended to patients with RCM to reduce risk of thromboembolic events.

Treating the associated systemic diseases and underlying conditions of RCM is important and in need. For example, patients with both PGM1 deficiency, a congenital glycosylation disorder, and RCM were treated with oral galactose treatment. Oral galactose did not benefit heart failure symptoms but improved the myopathy and exercise tolerance [57]. Amyloidosis, one of the infiltrative disorders linked to RCM, has had recent advances in treatment strategies. Genetic and molecular mechanism-oriented therapies are emerging for treatment of amyloidosis such as orally available transthyretin stabilizers, monoclonal antibodies and gene-editing strategies that have been effective in preclinical studies and successfully advanced in human clinical trials [58]. Approved for polyneuropathy in hereditary ATTR, patisiran, vutrisiran, and inotersen are short interfering RNA (siRNA) used to silence mutant and wild type TTR genes in order to halt neuropathy impairment and improve quality of life [59, 60, 61]. In 2019, the U.S. Food and Drug Administration (FDA) approved tafamidis, a selective transthyretin stabilizer, for the treatment of transthyretin amyloid cardiomyopathy (ATTR-CM). Tafamidis is not curative but has been shown to slow the progression of disease with reduced mortality and hospitalization [62, 63]. This is particularly true in less severe cases of ATTR-CM and early diagnosis [64]. Gene-editing approaches employ innovative technologies such as transthyretin-encoding gene silencing by RNA interference (RNAi) or antisense oligonucleotides and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 techniques [61, 65, 66].

Despite clinical management of heart failure symptoms and treatment of secondary RCM, progression to severe disease requires cardiac transplantation. Left ventricular assist device (LVAD) implantation is not commonly used in patients with RCM compared with DCM or ischemic cardiomyopathy patients [67]. A systematic review of six retrospective studies reported a higher risk of operative and short-term mortality in RCM patients with refractory heart failure undergoing LVAD implantation [68]. Prolonged LVAD implantation was pursued in two pediatric patients with pulmonary hypertension and was found to reduce pulmonary artery pressure and vascular resistance [69]. Durable mechanical circulatory support with biventricular assist device or total artificial heart has been considered for patients with amyloidosis who developed advanced heart failure [70]. Outcomes of adult RCM patients after heart transplantation have been comparable to those of non-RCM patients. However, increased mortality was observed in secondary RCM subgroup with amyloidosis and radiation/chemotherapy [71].

Pediatric patients have a waitlist mortality of around 10% at 1-year. Risk of death while on the waiting list increases with younger age, dependence on ventilator or multiple assist devices, or inotropic support. Survival rates for RCM patients 10 years after heart transplant is 63% and is comparable to non-cardiomyopathy patients and patients with other cardiomyopathies [72, 73]. Based on the PCMR database with 3375 cases of RCM between 1990–2008, 1-, 2-, and 5-year survival for RCM was 82%, 80%, and 68%, respectively [4]. Pediatric cases may result in sudden cardiac death (SCD) or complications from RCM, most commonly heart failure, arrhythmia, and ischemia-related complications. The poor clinical outcomes were due to ventricular arrhythmias, abnormal blood pressure in response to exercise, progressive elevation of pulmonary vascular resistance, and unexplained syncope [39, 74, 75]. Heart failure, lower fractional shortening z-score, and higher posterior wall thickness z-score at diagnosis are risk factors for survival without transplantation [4]. Clinical outcomes and mortality from RCM are worse in comparison to those in HCM and DCM, and survival is limited without cardiac transplantation [39, 76].

Molecular genetics has expanded the emerging potential to predict the progression of RCM and treatment strategies to prevent early mortality for inherited RCM [77]. Targeted therapies may be helpful in reversing pathogenic phenotypes or halting cardiac remodeling. For example, allele-specific silencing has been investigated in mice for the N47K mutation in the myosin light chain-2 (MYL2) gene. Injecting mice with RNAi showed the potential for a targeted therapy in RCM that partly regains normal function in diseased cells [78]. While CRISPR-Cas9 gene editing has not been studied in RCM, but it presents a potential additional therapeutic strategy and a technique to repair mutations or change gene expression in human induced pluripotent stem cell (hiPSC) [79, 80]. In 2017, Ma et al. [81] repaired a heterozygous dominant mutation of MYBC3, a mutation linked to familial HCM, in a human preimplantation embryo. MYBC3 mutations have also been seen in RCM patients; CRISPR-Cas9 gene editing studies are needed to determine its efficacy in RCM.

6. Histological changes in patients with RCM

Due to the broad group of mechanisms of pathogenesis in RCM, such as genetic, non-genetic, or secondary to a systemic disorder, histological changes in RCM are heterogeneous. On this basis, Sen-Chowdhry et al. [82] suggested RCM as a myocardial disorder in which restrictive ventricular physiology develops an early and dominant feature in the disease course. EMB has a high diagnostic yield in infiltrative disorders. Amyloidosis, sarcoidosis, hereditary hemochromatosis, and Fabry disease are identified by Congo red stain, granulomatosis reaction, iron, and ceramide trihexoside, respectively [25]. Additional techniques such as immunohistochemistry, Western blotting, mass spectrometry, amino acid sequencing, and electron microscopy are useful for amyloid fibril protein identification for an early diagnosis to prevent rapid disease deterioration [83, 84].

Autopsy studies of idiopathic RCM patients reveal ischemic damage preceding SCD [16]. In a cohort of seven patients diagnosed with RCM between 1974 and 1993, EMB revealed interstitial fibrosis [85]. A study by Kawano et al. [86] describes electron microscopy findings of a 65-year-old Japanese woman with a myosin heavy chain 7 (MYH7) causative mutation for RCM. Results from EMB of the left ventricle revealed focal myocyte necrosis, myofibrillar disarrangement, and scattered electron-dense material, some associated with Z-bands. Myofibrils without Z-bands were also identified, and tubular structures and pseudo-inclusion bodies were imaged in the nucleus [86]. Histological samples from a patient with a Q529X mutation in myopalladin (MYPN) revealed disorganized cardiomyocytes with bizarrely shaped nuclei, myofibrillar disarrangement, and hypertrophy. Side-branching and side-to-side junctions were observed in cardiomyocytes [87]. In biopsies from RCM patients with mutations in sarcomeric proteins, electron-dense material and Z-band disruption are the most common histopathological finding [86].

7. Molecular genetics of primary RCM

Approximately 30% of RCM cases have a family history of RCM considered as a primary disease of genetic origin, but only a few genes, such as cardiac troponin T (cTnT) and troponin I (cTnI), myosin-binding protein C (MYBPC), MYH7, myosin light chain-2 and 3 (MYL2, MYL3), desmin (DES), MYPN, titin (TTN), Bcl2-associated athanogene 3 (BAG3), discoidin, CUB and LCCL domain containing 2 (DCBLD2), lamin A/C (LMNA), and filamin C (FLNC) have been associated with familial RCM [87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105] (Table 2, Ref. [87, 89, 97, 98, 102, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126]).

Table 2.Mutations associated with RCM in humans that were studies in animals for modeling RCM.
Gene Protein Human mutation Animal mutation models Pathogenesis
BAG3 Bcl2-associated athanogene 3 P209L P209L overexpression [102] Autophagy, Z-disc disruption, protein aggregates, and increased cardiac fibroblasts.
DCBLD2 Discodin, CUB, LCCL domain-containing protein 2 W27* Dcbld2 KO [106] Impaired normal vascular development via altered VEGF signaling.
DES Desmin R406W R405W Desmin aggregates and disruption of intercalated discs.
KI [107]
FLNC Filamin C S1624L A1186V Myotube defects, decreased sarcomere localization, protein aggregates, Z-disc disruption, autophagy.
I2160F [97] A1183L
V2297M [108] zebrafish overexpression
A1186V
A1183L [109]
P2298L [110]
T2563C
T2710X [110]
LMNA Lamin A/C 835delG [104] N/A Not known. Likely protein truncation or loss of protein via nonsense‐mediated mRNA decay.
MYBPC-3 Myosin-binding protein C Ala1105Glyfs*85 [105] N/A Not known. Likely protein truncation or loss of protein via nonsense‐mediated mRNA decay.
MYH7 β-myosin heavy chain G768R [111] N/A Not known.
P838L [112]
MYL2 Myosin light chain 2 N47K [113] N47K Tg Silencing of the 47K allele reduced cardiac fibrosis, LV mass, hypertrophic biomarkers, and increased survival.
N47K AAV9-RNAi [78]
MYL3 Myosin light chain 3 E143K [114] E143K [115] Hypercontractility, increased binding affinity to actin, and slow dissociation of acto-myosin complex.
MYPN Myopalladin Q529X [87] Q526X KI [116] Reduced levels of CARP, Mek1/2, Erk1/2, Smad2 and Akt.
TNNI3 Cardiac troponin I L144Q [117] Increased Ca2+ sensitivity, ATPase activity, and force generation; exposed myosin-binding sites.
R145W R145W [118, 119]
R170W
A171T
K178E [120] K178E [121, 120, 122]
D190G
D190H
R192H R193H [123]
R192C [124]
TNNT2 Troponin T type 2 E136K [89] Drosophila melanogaster E87K Increased resting tension and reduced sarcomere length.
I79N [125] Rat E138K
I79N Tg [126]
TTN Titin Y7621C [98] N/A Myocyte degeneration, Z-disc disruption, and fibrosis.
Comment: All mutations are studied in mouse models unless indicated otherwise.

The genetic mutations associated with inherited RCM are primarily in structural proteins of the sarcomere or proteins associated with the sarcomere [77]. The sarcomere is the contractile unit of skeletal and cardiac muscle with thin, thick, and elastic filaments anchored by the M-line and Z-disc [127]. The thick filaments are made up of myosin and myosin-binding protein C, a regulatory protein at the C region cross-bridging zone, and thin filaments are comprised of actin and regulatory proteins troponin and tropomyosin. At low calcium concentrations, troponin and tropomyosin bind and inhibit the myosin ATPase and force generation. Calcium release from the sarcoplasmic reticulum causes the sarcomere to contract from thick and thin filaments sliding past one another [128]. Contractile function relies on a delicate balance between the filaments and troponin complexes to control the state of the sarcomere involved in homeostasis of cardiac function [129]. Most of the RCM cases with genetic variants involved directly or indirectly in sarcomere function have been reported to cause childhood RCM or cases that rapidly deteriorate due to conduction defects and cardiac restrictive dysfunction necessitating heart transplantation [97, 98, 103, 104, 124, 130].

Several mutations found from family studies have been used in animal studies of RCM as shown in Table 2. Animal models with analogous human mutations in myosin, cTnI, MYPN, and DES yield a RCM phenotype of diastolic dysfunction [131]. Cells from patients with cardiomyopathies can be taken to develop hiPSC lines for cell studies on cardiomyocytes to test therapeutics and detect cardiotoxicity [79]. Using CRISPR-Cas9 gene editing, researchers can create isogenic variants of hiPSC lines to identify cardiotoxic mutations and investigate a pathogenic mutation’s effect on physiology, contractility, and morphology [132]. In addition, single short guide RNAs through CRISPR-Cas9 editing has been shown to disrupt genes Myh6, Sav1, and Tbx20 in postnatal mice [133]. With the hope of developing a human RCM therapy, the adeno-associated virus (AAV) - RNAi approach showed the feasibility, efficacy, and safety of preclinical therapeutics in MYL2 N47K mutation transgenic mice [78].

Some of the examples of clinical or histopathological changes in RCM patients with genetic mutations listed in Table 2 may reveal features of HCM, suggesting that both diseases may share similar pathogenic and genetic mechanisms [134]. While HCM and RCM are phenotypically different, both are considered disorders of diastolic dysfunction. Vio et al. [35] suggested that familial RCM cases may represent a part of the phenotypic spectrum of HCM rather than a different genetic cardiomyopathy. Differential diagnosis between primary RCM and HCM can be difficult when ventricular hypertrophy is mild. Kubo et al. [135] distinguished the “restrictive phenotype” as a severe diastolic dysfunction with restrictive filling that is in the clinical spectrum of HCM with no or minimal left ventricular hypertrophy of 15 mm. In this large study involving 1226 patients with HCM, the “restrictive phenotype” was associated with severe functional limitation and poor prognosis. Variants in MYH7 and cTnI were determined as the underlying origin [135]. In another study, cTnI was the causative gene of idiopathic RCM and HCM diagnoses in a large family [41]. Mogensen et al. [117] reported on a male teenage patient diagnosed with RCM and with the de novo mutation R192H. Echocardiography revealed bi-atrial enlargement, normal left ventricular wall thickness, and normal systolic function, consistent with the RCM diagnosis; however, postmortem microscopy of heart tissue revealed myocyte disarray and cardiomyocyte hypertrophy which is characteristic of HCM. In cTnI positive cases, mutations have been found to be associated with either HCM or RCM depending on the specific amino acid substitution. For example, R145G and R145Q are linked to HCM while R145W is associated with RCM [136, 137]. More recently, a frame-shift variant p.Ala1105Glyfs*85 in MYBPC3 has been associated to both RCM and HCM in a Korean family [105]. Proband displayed normal LV chamber size, borderline LV hypertrophy and severe biatrial enlargement, while his affected daughters had asymmetric septal hypertrophy of the LV consistent with HCM. Hence, even in familial cases of cardiomyopathy, mutations in the same gene or same mutation may yield a range of clinical phenotypes and symptoms.

Beyond genetic mutations of genes expressed in cardiac myocytes, cardiac fibroblasts and the extracellular matrix (ECM) may be involved with the pathogenesis of RCM [138]. Natural feline RCM is characterized by severe and extensive fibrous thickening of the endocardium and chamber deformity [139]. Despite these animal models and cell lines elucidating the pathogenesis and pathophysiology as well genetic therapies, primary RCM remains one of the least studied and understood cardiomyopathies. The following sections will outline the genetic variants linked to RCM and the role of paracrine factors secreted by cardiac fibroblasts.

7.1 Cardiac troponin I and T and calcium sensitivity

Troponin I (cTnI) inhibits sarcomere contraction by binding to the Ca2+ binding subunit of cardiac troponin C (cTnC) and cardiac troponin T (cTnT) [129, 140]. Mutations in cTnI make up most of the genetic mutations linked to RCM [141]. Six human cTnI missense mutations were first isolated in 2003 in a single large family study [117]. The mutations, L144Q, R145W, A171T, K178E, D190G, and R192H, are located in the C-terminal domain, a conserved portion which encodes for actin-filament binding sites and regulates calcium interactions with troponin C. Mutations K178E and R192H are associated with severe clinical phenotypes of RCM in humans and in transgenic mice with corresponding mutations, K178E and R193H, which exhibit increased calcium sensitivity, diastolic dysfunction, and atrial enlargement [1, 142]. The K178E mutation induced early mortality in transgenic mice, and mice exhibited increased resting tension and decreased compliance [143]. In R193H mutant mice, transgenic cTnI displays increased binding affinity to histone deacetylase 1 (HDAC1). Increased expression of HDAC1 downregulates phosphodiesterase 4D (PDED4D). Epigallocatechin gallate (EGCG), which reduces Ca2+ sensitivity, is a HDAC1-inhibitor and prevents downregulated PDE4D in R193H mice [144]. The p.R192C mutation has been identified in twin sisters with RCM and linked to a poor prognosis [145].

Transgenic R145W mice have also been studied as a model for RCM. Papillary fibers in mutant mice demonstrate increased Ca2+ sensitivity for force generation and ATPase activity due to structural change in mutant cTnI. The R145W mutant mouse demonstrates prolonged force and intracellular Ca2+ transients which likely is responsible for diastolic dysfunction due to resistance of ventricular filling. Wen et al. [118] hypothesized that the disruption in calcium homeostasis affects the cell-to-cell signaling of gap junctions. A study with multicellular skinned human myocardium found that the R145W mutation affects the interaction between helix C of cTnC with cTnI, leading to increased Ca2+ sensitivity and uncoupling from protein kinase A (PKA) phosphorylation [146], while a similar mutation R145G causes modulation of troponin structure by PKA phosphorylation, resulting in HCM [119].

Certain amino acid substitutions can have varying effects depending on the charge differences. Removing or substituting charged residues on tropomyosin-binding sites impairs normal function of cTnT and relaxation [147]. Equivalent E136K mutations were studied in Drosophila melanogaster (E87K) and in rats (E138K). Mutant E87K Drosophila demonstrate increased resting tension and slight reduction in sarcomere length. The rat E138K mutation showed similar results with increased resting tension, reduced sarcomere lengths, greater contractility, and prolonged relaxation periods [147].

7.2 Cardiac myosin light and heavy chains

Myosin is a protein complex comprised of two heavy chains - α-myosin heavy chain (MYH6) and β-myosin heavy chain (MYH7) — and four light chains — two regulatory light chains (MYL2) and two essential light chains (MYL3). The E143K mutation in myosin essential light chain, encoded by MYL3, has been linked to clinical phenotypes of HCM with restrictive physiology, severe pediatric HCM, and RCM with LV hypertrophy [148, 149]. Typically, homozygous 143K patients display the RCM phenotype. While MYL3 mutations have been studied in HCM murine models, Yuan et al. [115] used transgenic mice with the E143K mutation as a model for RCM. The charge difference due to the amino acid substitution affected actin-myosin interactions, leading to hypercontractility from increased binding affinity to actin, actin-activated ATPase activity, and slower dissociation of the actin-myosin complex. Myosin light chain phosphorylation, which is linked to abnormal cardiac phenotypes, is decreased in E143K mice while cTnI phosphorylation remained unchanged. Collagen type-I and III and stress response genes were upregulated, contributing to the structural abnormalities and cardiac remodeling of the E143K mice.

Allele-specific silencing, using AAV-RNAi (AAV9-M7.8L), was found to reduce mutant allele expression, hypertrophic biomarkers, and intramyocardial fibrosis in mice with the human RCM-analogous mutation N47K. In single cell studies, cardiac myocytes treated with RNAi were shown to partially restore normal contraction, relaxation, and calcium reuptake [78]. Although this therapeutic strategy has only been conducted in animal studies, RNAi therapies may be an effective strategy to reverse pathogenic phenotypes.

Myosin heavy chain, along with myosin binding protein C, are the genes most affected by HCM. The RCM mutation G768R in exon 21 in the MYH7 was described in a 15-month-old infant presenting with a heart murmur, gallop, and diastolic dysfunction [95].

7.3 Titin

Titin, a giant filament protein, is frequently mutated in cardiomyopathies and phosphorylation of titin affects passive tension, Ca2+ sensitivity, and force development [128]. Titin connects the Z-disc to the M-line and is involved with signal transduction. Titin acts like a spring and regulates resting tension and elasticity [150]. De novo mutation Y7621C in the fibronectin-3 domain, a highly conserved region of the TTN gene, was identified in a family study. Endomyocardial biopsy revealed myocyte degeneration and fibrosis [98].

7.4 Filamin C

Filamins A, B, and C cross-link actin rods at the Z-disc of sarcomeres, and FLNC is primarily expressed in cardiac and skeletal muscle, which explains the pleiotropic effects of its genetic mutations in cardiomyopathies and myopathies [151, 152]. Mutations S1624L, V2297M, and I2160F have been associated with RCM, and it is hypothesized that loss-of-function of the gene contributes to pathogenesis [108]. Histology of heart tissue of patients with S1624L and I2160F revealed eosinophilic cytoplasmic inclusions and filament accumulation, suggestive of protein aggregation [97]. The mutation V2297M was identified as the pathogenic mutation in a pedigree with phenotypes of RCM, atrial fibrillation, ventricular tachycardia, and congenital heart defects. Histology of the proband’s heart revealed interstitial fibrosis, myocyte hypertrophy, and mild atrial amyloid deposition, and the pathology report of the proband’s sister, also diagnosed with an RCM phenotype, identified severe interstitial fibrosis and myocyte hypertrophy. In bi-allelic CRISPR-Cas9 insertions of V2297M in human embryonic stem cells, contractile function was diminished with no changes in FLNC expression [108]. Recently, the P2298L, T2563C and T2710X variants have been reported in pediatric RCM [110].

7.5 Lamin A/C

Mutations in LMNA, an intermediate protein meshwork lining the nucleoplasmic face of the inner nuclear membrane, are mostly associated with DCM, metabolic diseases, conduction disorders, and neuromuscular disorders [153, 154]. The heterozygous 835delG mutation was shown to be associated with RCM in a 53-year-old man with exertional dyspnea who received a cardiac transplant two years after diagnosis. His sister presented with the same mutation; however, she was asymptomatic [155].

7.6 Myosin-binding protein C

Myosin-binding protein C is a myosin-associated protein found in the cross-bridge-bearing zone (C region) of A-bands in striated muscle [156]. Mutations in MYBPC are common in HCM and less common in RCM. The frameshift mutation Ala1105Glyfs*85 was identified as the causative mutation of RCM in a 74-year-old Korean male with exertional dyspnea, palpitations, and pitting edema in the lower extremities. The patient’s two children were diagnosed with HCM, speaking to the pleiotropic nature of this mutation and probable impact of modifier genes and environmental factors [105].

7.7 Myopalladin

Myopalladin, a 145-kDa protein with five immunoglobulin domains, is encoded by MYPN gene located on chromosome 10q21.3 [157]. The protein interacts with cardiac ankyrin repeat protein (CARP/ANKRD1) to regulate Z-disc stretch-signaling to the nucleus of cardiomyocytes. In family studies, MYPN heterozygous mutations have been associated with HCM, DCM, and RCM [87]. Using one such human Q529X mutation, heterozygote knock-in mice and homozygote mice carried the corresponding murine MYPN mutation (Q526X) in exon 10 as a model for RCM [158]. At 12 weeks of age, heterozygote MypnWT/Q526X mice retained systolic function and exhibited impaired diastolic filling in the left ventricle with decreased end-diastolic volume in comparison to wildtype and homozygote mice (Fig. 3). The E/A ratio of the mitral valve was significantly increased in heterozygotes compared with wildtype mice. Histologically, no cardiomyocyte hypertrophy was found in heterozygotes. The mice displayed fibrotic remodeling, specifically interstitial and perivascular fibrosis in ventricular myocardium. Mutant Mypn protein translocates to the nucleus where it may affect levels of CARP. Total and nuclear CARP levels and phosphorylation of Mek1/2, Erk1/2, Smad2 and Akt were reduced in MypnWT/Q526X mice. Markers of heart failure (Nppa, Nppb, and Myh6) and fibrosis (Tgfβ1,αSma, Opn, and Postn) were upregulated [116]. Mutant human MYPN Q529X was not able to translocate to the nucleus in vitro, while CARP and wildtype MYPN had a complete translocation. This suggests that MYPN-CARP interactions are not necessary for MYPN translocation [158, 159].

Gu et al. [160] applied a systems genetics approach to investigate the regulation of MYPN for discovery its gene network involved in RCM. Gene expression values were measured in the heart of a large family of BXD recombinant inbred (RI) mice derived from C57BL/6J and DBA/2J, and expression quantitative trait locus (eQTL) mapping and gene enrichment analysis results were validated on proteomic levels in Mypn knock-in mice. Two novel modifier genes, Syne1 and Myom1, have been confirmed at the protein level that could be involved in RCM development.

7.8 BAG3

BAG3 is an anti-apoptotic protein that also plays a role in mediating autophagy and targeting aggregated proteins for degradation [161]. Mutation P209L causes severe pediatric RCM, muscular dystrophy, and respiratory distress. The mutation is located at the heat shock protein (HSP) 8 binding site; however, it does not affect binding of HSP8 - instead, it promotes aggregation [102]. Interestingly, murine models have yielded various phenotypes depending on the study. The murine equivalent mutation, P215L, did not lead to cardiomyopathy in knock-in mice for up to 16 months. Physiology and morphology of mutant hearts revealed no changes from wildtype hearts, and measurements of ubiquitinated proteins, Hsps, and autophagy were unaffected [162]. On the other hand, cardiomyocyte-specific αMHC-human Bag3 P209L transgenic mice displayed heart failure, pre-amyloid oligomers at 12 months, and increased activated cardiac fibroblasts with no change in fibrosis [163]. Recently, Kimura et al. [102] reported overexpression of P209L in transgenic mice causes disruption in Z-disc integrity and aggregate formation. Disruption in the HSP8 binding site leads to increased autophagy in cardiac tissue [102].

7.9 Desmin

Desmin is a muscle-specific intermediate filament that integrates the sarcomere, Z-discs, nuclear membrane, and intercalated discs acting as a structural component as well as a regulator of the sarcomeric architecture [164]. A mutation in DES, R406W, was identified in a young male patient as the causative gene of progressive RCM. Mutated desmin filaments can integrate with wildtype filaments; however, under mechanical stress, aggregates will form. The R406W mutation did not allow desmin filaments to attach to intercalated discs and affected organization of spectrin proteins, N-cadherin, and connexin-43. Disruption in the intercalated discs may produce conduction abnormalities and arrhythmias, a common clinical manifestation of RCM [165]. The R406W mutation was recapitulated in knock-in heterozygous and homozygous mice with an analogous murine mutation, Des R405W. Histology demonstrated similar pathology with desmin aggregates and misalignment of myofibrils. Homozygous mutant mice died within the first three months of life from complications in gut smooth muscle [107].

7.10 DCBLD2

A DCBLD2 gene variant has recently been identified in a 5-year-old patient with RCM, developmental delay, and dysmorphic features. The DCBLD2 gene encodes a type-I transmembrane receptor involved with intercellular signaling, cell growth regulation, and wound healing. Heterozygous V429L variant in ACVRL1 and homozygous nonsense mutation W27* in DCBLD2 were identified in the patient, although the DCBLD2 was determined as the causative mutation. Angiogenesis was reduced in knockout DCBLD2 mice [166].

7.11 Paracrine signaling and extracellular influence on pathogenesis

Impaired ventricular filling and fibrosis, hallmarks of RCM, was evaluated by Tsuru et al. [139] to determine how paracrine factors secreted by cardiac fibroblasts may impact cardiomyocyte dysfunction, causing the pathogenic phenotype. Under mechanical stress and ischemic damage, quiescent cardiac fibroblasts differentiate into myofibroblasts and secrete ECM proteins and cytokines. Cardiac fibroblasts are the most common cells found in the heart and play a significant role in ischemic damage and pathogenic hemodynamics. Myofibroblasts participate in scar formation and play a role in ischemic damage, expressing α-smooth muscle actin (α-SMA) and increasing contractility. In contrast, fibroblasts seen in RCM have normal α-SMA expression and low contractility, while maintaining secretion of ECM proteins and cytokines [167]. Cell lines were cultured from human cardiac fibroblasts, taken from four pediatric RCM patients - three with TNNI3 mutations (K178E, R170W, R192H) and one with no detectable causative mutation. Fibroblasts were grown in direct and indirect co-culture with healthy cardiomyocytes, causing a decrease in relaxation velocity in the cardiomyocytes. Gene expression for ECM components, ITGA11, COL16A1, TGFB1, MMP2, and COL1A1, were upregulated in fibroblasts from the RCM patients, while cytokines FGF5, CXCL12, MMP1, and IL33 were downregulated, suggesting that dysregulation of paracrine factors may be involved with the diastolic dysfunction found in RCM [138].

8. Conclusion and future directions

RCM is pleiotropic and, in some family studies, the genotype-phenotype correlation can be ambiguous due to the complex genetic network and influence from environmental factors and modifier genes. Family studies, human cell lines, animal models, and systems genetics analysis are important tools for determining disease-causing variants of RCM and elucidating the pathogenic mechanism(s). Cardiomyopathies overlap in causative mutations and associated genes. Hence, there is a pressing need for genetic testing in family members of patients with cardiomyopathy. Whole exome sequencing and next generation sequencing are important techniques for identifying gene variants. Proteins encoded by BAG3, DES, and FLNC exhibit histopathological protein aggregation, and sarcomeric proteins cTnI and titin affect Ca2+ sensitivity. Recently, researchers have delved into cell lines separate from cardiomyocytes, specifically cardiac fibroblasts and the ECM. Paracrine factors may negatively influence healthy cardiomyocytes to develop abnormal relaxation.eis While genes linked to RCM share some similar cardiotoxic effects on cardiomyocytes, multiple mechanisms may lead to the RCM phenotype. Genetic studies present an opportunity to pinpoint targeted therapy that can reverse pathogenic abnormalities of RCM.

Author contributions

MC—Data curation, Investigation, Project administration, Writing - review and editing; BOO, NRA—Data curation, Investigation, Writing - review & editing; JAT—Funding acquisition, Resources, Writing - review & editing; EP—Conceptualization, Methodology, Writing - original draft, Writing - review & editing. All authors have read and agreed to the published version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

Thanks to all the peer reviewers for their opinions and suggestions.

Funding

The study was supported in part by National Institutes of Health [R01HL53392 and R01HL116906 (Towbin) and R01HL151438 (Multiple PI: Towbin, Purevjav, Lu)].

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Dadson K, Hauck L, Billia F. Molecular mechanisms in cardiomyopathy. Clinical Science. 2017; 131: 1375–1392.
[2]
Elliott P, Andersson B, Arbustini E, Bilinska Z, Cecchi F, Charron P, et al. Classification of the cardiomyopathies: a position statement from the european society of cardiology working group on myocardial and pericardial diseases. European Heart Journal. 2007; 29: 270–276.
[3]
Muchtar E, Blauwet LA, Gertz MA. Restrictive Cardiomyopathy : Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circulation Research. 2017; 121: 819–837.
[4]
Webber SA, Lipshultz SE, Sleeper LA, Lu M, Wilkinson JD, Addonizio LJ, et al. Outcomes of restrictive cardiomyopathy in childhood and the influence of phenotype: a report from the Pediatric Cardiomyopathy Registry. Circulation. 2012; 126: 1237–1244.
[5]
Russo LM, Webber SA. Idiopathic restrictive cardiomyopathy in children. Heart. 2005; 91: 1199–1202.
[6]
BRIGDEN W. Uncommon myocardial diseases: the non-coronary cardiomyopathies. Lancet. 1957; 273: 1243–1249.
[7]
TEARE D. Asymmetrical hypertrophy of the heart in young adults. British Heart Journal. 1958; 20: 1–8.
[8]
Hollman A, Goodwin JF, Teare D, Renwick JW. A family with obstructive cardiomyopathy (asymmetrical hypertrophy). British Heart Journal. 1960; 22: 449–456.
[9]
Goodwin JF, Gordon H, Hollman A, Bishop MB. Clinical aspects of cardiomyopathy. British Medical Journal. 1961; 1: 69–79.
[10]
Goodwin JF, Oakley CM. The cardiomyopathies. British Heart Journal. 1972; 34: 545–552.
[11]
Chew CY, Ziady GM, Raphael MJ, Nellen M, Oakley CM. Primary restrictive cardiomyopathy. Non-tropical endomyocardial fibrosis and hypereosinophilic heart disease. British Heart Journal. 1977; 39: 399–413.
[12]
Report of the WHO/ISFC task force on the definition and classification of cardiomyopathies. British Heart Journal. 1980; 44: 672–673.
[13]
Richardson P, McKenna W, Bristow M, Maisch B, Mautner B, O’Connell J, et al. Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Definition and Classification of cardiomyopathies. Circulation. 1996; 93: 841–842.
[14]
Kostareva A, Kiselev A, Gudkova A, Frishman G, Ruepp A, Frishman D, et al. Genetic Spectrum of Idiopathic Restrictive Cardiomyopathy Uncovered by Next-Generation Sequencing. PLoS ONE. 2016; 11: e0163362.
[15]
Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, et al. Contemporary Definitions and Classification of the Cardiomyopathies. Circulation. 2006; 113: 1807–1816.
[16]
Albakri A. Restrictive cardiomyopathy: A review of literature on clinical status and meta-analysis of diagnosis and clinical management methods table of contents. Internal Medicine and Care. 2018; 2: 1–15.
[17]
Pereira NL, Grogan M, Dec GW. Spectrum of Restrictive and Infiltrative Cardiomyopathies: Part 1 of a 2-Part Series. Journal of the American College of Cardiology. 2018; 71: 1130–1148.
[18]
Felker GM, Thompson RE, Hare JM, Hruban RH, Clemetson DE, Howard DL, et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. The New England Journal of Medicine. 2000; 342: 1077–1084.
[19]
Nugent AW, Daubeney PEF, Chondros P, Carlin JB, Cheung M, Wilkinson LC, et al. The epidemiology of childhood cardiomyopathy in Australia. The New England Journal of Medicine. 2003; 348: 1639–1646.
[20]
Kapoor M, Das S, Biswas A, Seth S, Bhargava B, Rao VR. Epidemiology of cardiomyopathy — A Clinical and Genetic Study of Restrictive Cardiomyopathy: The EPOCH-R Study. Journal of the Practice of Cardiovascular Sciences. 2017; 3: 143–149.
[21]
Garcia-Pavia P, Rapezzi C, Adler Y, Arad M, Basso C, Brucato A, et al. Diagnosis and treatment of cardiac amyloidosis. A position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. European Journal of Heart Failure. 2021; 23: 512–526.
[22]
Shah KB, Inoue Y, Mehra MR. Amyloidosis and the heart: a comprehensive review. Archives of Internal Medicine. 2006; 166: 1805–1813.
[23]
Kyriakou P, Mouselimis D, Tsarouchas A, Rigopoulos A, Bakogiannis C, Noutsias M, et al. Diagnosis of cardiac amyloidosis: a systematic review on the role of imaging and biomarkers. BMC Cardiovascular Disorders. 2018; 18: 221.
[24]
Falk RH. Diagnosis and management of the cardiac amyloidoses. Circulation. 2005; 112: 2047–2060.
[25]
Brown KN, Pendela VS, Diaz RR. Restrictive Cardiomyopathy, in StatPearls. StatPearls Publishing. StatPearls Publishing LLC.: Treasure Island (FL). 2021.
[26]
Mohammed SF, Mirzoyev SA, Edwards WD, Dogan A, Grogan DR, Dunlay SM, et al. Left ventricular amyloid deposition in patients with heart failure and preserved ejection fraction. JACC: Heart Failure. 2014; 2: 113–122.
[27]
Teng C, Li P, Bae JY, Pan S, Dixon RAF, Liu Q. Diagnosis and treatment of transthyretin‐related amyloidosis cardiomyopathy. Clinical Cardiology. 2020; 43: 1223–1231.
[28]
Markatis E, Afthinos A, Antonakis E, Papanikolaou IC. Cardiac sarcoidosis: diagnosis and management. Reviews in Cardiovascular Medicine. 2020; 21: 321.
[29]
Morimoto T, Azuma A, Abe S, Usuki J, Kudoh S, Sugisaki K, et al. Epidemiology of sarcoidosis in Japan. European Respiratory Journal 2008; 31: 372–379.
[30]
Dubrey SW, Bell A, Mittal TK. Sarcoid heart disease. Postgraduate Medical Journal. 2007; 83: 618–623.
[31]
Bresnitz EA, Strom BL. Epidemiology of sarcoidosis. Epidemiologic Reviews. 1983; 5: 124–156.
[32]
Dumas O, Abramovitz L, Wiley AS, Cozier YC, Camargo CA. Epidemiology of Sarcoidosis in a Prospective Cohort Study of U.S. Women. Annals of the American Thoracic Society. 2016; 13: 67–71.
[33]
Baughman RP, Field S, Costabel U, Crystal RG, Culver DA, Drent M, et al. Sarcoidosis in America. Analysis Based on Health Care Use. Annals of the American Thoracic Society. 2016; 13: 1244–1252.
[34]
Hulten E, Aslam S, Osborne M, Abbasi S, Bittencourt MS, Blankstein R. Cardiac sarcoidosis-state of the art review. Cardiovascular Diagnosis and Therapy. 2016; 6: 50–63.
[35]
Vio R, Angelini A, Basso C, Cipriani A, Zorzi A, Melacini P, et al. Hypertrophic Cardiomyopathy and Primary Restrictive Cardiomyopathy: Similarities, Differences and Phenocopies. Journal of Clinical Medicine. 2021; 10: 1954.
[36]
Lee TM, Hsu DT, Kantor P, Towbin JA, Ware SM, Colan SD, et al. Pediatric Cardiomyopathies. Circulation Research. 2017; 121: 855–873.
[37]
Lipshultz SE, Cochran TR, Briston DA, Brown SR, Sambatakos PJ, Miller TL, et al. Pediatric cardiomyopathies: causes, epidemiology, clinical course, preventive strategies and therapies. Future Cardiol. 2013; 9(6): 817–848.
[38]
Denfield SW, Rosenthal G, Gajarski RJ, Bricker JT, Schowengerdt KO, Price JK, et al. Restrictive cardiomyopathies in childhood. Etiologies and natural history. Texas Heart Institute Journal. 1997; 24: 38–44.
[39]
Rivenes SM, Kearney DL, Smith EO, Towbin JA, Denfield SW. Sudden death and cardiovascular collapse in children with restrictive cardiomyopathy. Circulation. 2000; 102: 876–882.
[40]
Rammos A, Meladinis V, Vovas G, Patsouras D. Restrictive Cardiomyopathies: the Importance of Noninvasive Cardiac Imaging Modalities in Diagnosis and Treatment-a Systematic Review. Radiology Research and Practice. 2017; 2017: 2874902.
[41]
Mogensen J, Arbustini E. Restrictive cardiomyopathy. Current Opinion in Cardiology. 2009; 24: 214–220.
[42]
Ammash NM, Seward JB, Bailey KR, Edwards WD, Tajik AJ. Clinical profile and outcome of idiopathic restrictive cardiomyopathy. Circulation. 2000; 101: 2490–2496.
[43]
Leya FS, Arab D, Joyal D. The Efficacy of Brain Natriuretic Peptide Levels in Differentiating Constrictive Pericarditis from Restrictive Cardiomyopathy. ACC Current Journal Review. 2005; 14: 38.
[44]
Dispenzieri A, Gertz MA, Kyle RA, Lacy MQ, Burritt MF, Therneau TM, et al. Serum cardiac troponins and N-terminal pro-brain natriuretic peptide: a staging system for primary systemic amyloidosis. Journal of Clinical Oncology. 2004; 22: 3751–3757.
[45]
Sanchorawala V. Light-chain (AL) amyloidosis: diagnosis and treatment. Clinical Journal of the American Society of Nephrology. 2006; 1: 1331–1341.
[46]
Selvaganesh M, Arul AS, Balasubramanian S, Ganesan N, Naina Mohammed S, Sivakumar GS, et al. An unusual ECG pattern in restrictive cardimyopathy. Indian Heart Journal. 2015; 67: 362–367.
[47]
Mussinelli R, Salinaro F, Alogna A, Boldrini M, Raimondi A, Musca F, et al. Diagnostic and prognostic value of low QRS voltages in cardiac AL amyloidosis. Annals of Noninvasive Electrocardiology. 2013; 18: 271–280.
[48]
Oerlemans MIFJ, Rutten KHG, Minnema MC, Raymakers RAP, Asselbergs FW, de Jonge N. Cardiac amyloidosis: the need for early diagnosis. Netherlands Heart Journal. 2019; 27: 525–536.
[49]
Nagueh SF, Smiseth OA, Appleton CP, Byrd BF, Dokainish H, Edvardsen T, et al. Recommendations for the Evaluation of Left Ventricular Diastolic Function by Echocardiography: an Update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Journal of the American Society of Echocardiography. 2016; 29: 277–314.
[50]
Mahmoud A, Bansal M, Sengupta PP. New Cardiac Imaging Algorithms to Diagnose Constrictive Pericarditis Versus Restrictive Cardiomyopathy. Current Cardiology Reports. 2017; 19: 43.
[51]
Abduch MCD, Alencar AM, Mathias Jr. W, Vieira MLDC. Cardiac Mechanics Evaluated by Speckle Tracking Echocardiography. Arquivos Brasileiros De Cardiologia. 2014; 102: 403–412.
[52]
Lipshultz SE, Sleeper LA, Towbin JA, Lowe AM, Orav EJ, Cox GF, et al. The incidence of pediatric cardiomyopathy in two regions of the United States. The New England Journal of Medicine. 2003; 348: 1647–1655.
[53]
Parrott A, Khoury PR, Shikany AR, Lorts A, Villa CR, Miller EM. Investigation of de novo variation in pediatric cardiomyopathy. American Journal of Medical Genetics Part C: Seminars in Medical Genetics. 2020; 184: 116–123.
[54]
Heliö T, Elliott P, Koskenvuo JW, Gimeno JR, Tavazzi L, Tendera M, et al. ESC EORP Cardiomyopathy Registry: real‐life practice of genetic counselling and testing in adult cardiomyopathy patients. ESC Heart Failure. 2020; 7: 3013–3021.
[55]
Barry D, Herskovitz S, Agarwal S, Karr S. (10) Restrictive Cardiomyopathy in Toddler Aged Female. Pediatrics. 2018; 142: 387–394.
[56]
Mizia‐Stec K, Caforio ALP, Charron P, Gimeno JR, Elliott P, Kaski JP, et al. Atrial fibrillation, anticoagulation management and risk of stroke in the Cardiomyopathy/Myocarditis registry of the EURObservational Research Programme of the European Society of Cardiology. ESC Heart Failure. 2020; 7: 3601–3609.
[57]
Donoghue SE, White SM, Tan TY, Kowalski R, Morava E, Yaplito-Lee J. Galactose treatment of a PGM1 patient presenting with restrictive cardiomyopathy. JIMD Reports. 2020; 57: 29–37.
[58]
Griffin JM, Rosenblum H, Maurer MS. Pathophysiology and Therapeutic Approaches to Cardiac Amyloidosis. Circulation Research. 2021; 128: 1554–1575.
[59]
Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. New England Journal of Medicine. 2018; 379: 11–21.
[60]
Habtemariam BA, Karsten V, Attarwala H, Goel V, Melch M, Clausen VA, et al. Single‐Dose Pharmacokinetics and Pharmacodynamics of Transthyretin Targeting N‐acetylgalactosamine–Small Interfering Ribonucleic Acid Conjugate, Vutrisiran, in Healthy Subjects. Clinical Pharmacology & Therapeutics. 2021; 109: 372–382.
[61]
Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis. New England Journal of Medicine. 2018; 379: 22–31.
[62]
Rapezzi C, Elliott P, Damy T, Nativi-Nicolau J, Berk JL, Velazquez EJ, et al. Efficacy of Tafamidis in Patients with Hereditary and Wild-Type Transthyretin Amyloid Cardiomyopathy. JACC: Heart Failure. 2021; 9: 115–123.
[63]
Damy T, Garcia-Pavia P, Hanna M, Judge DP, Merlini G, Gundapaneni B, et al. Efficacy and safety of tafamidis doses in the Tafamidis in Transthyretin Cardiomyopathy Clinical Trial ( ATTR‐ACT ) and long‐term extension study. European Journal of Heart Failure. 2021; 23: 277–285.
[64]
Sultan MB, Gundapaneni B, Schumacher J, Schwartz JH. Treatment with Tafamidis Slows Disease Progression in Early-Stage Transthyretin Cardiomyopathy. Clinical Medicine Insights: Cardiology. 2017; 11: 117954681773032.
[65]
Suhr OB, Coelho T, Buades J, Pouget J, Conceicao I, Berk J, et al. Efficacy and safety of patisiran for familial amyloidotic polyneuropathy: a phase II multi-dose study. Orphanet Journal of Rare Diseases. 2015; 10: 109.
[66]
Finn JD, Smith AR, Patel MC, Shaw L, Youniss MR, van Heteren J, et al. A Single Administration of CRISPR/Cas9 Lipid Nanoparticles Achieves Robust and Persistent in Vivo Genome Editing. Cell Reports. 2018; 22: 2227–2235.
[67]
Sajgalik P, Grupper A, Edwards BS, Kushwaha SS, Stulak JM, Joyce DL, et al. Current Status of Left Ventricular Assist Device Therapy. Mayo Clinic Proceedings. 2016; 91: 927–940.
[68]
Sreenivasan J, Kaul R, Khan MS, Ranka S, Demmer RT, Yuzefpolskaya M, et al. Left Ventricular Assist Device Implantation in Hypertrophic and Restrictive Cardiomyopathy: a Systematic Review. ASAIO Journal. 2021; 67: 239–244.
[69]
Schlein J, Riebandt J, Laufer G, Zimpfer D. Reversal of pulmonary hypertension in paediatric patients with restrictive cardiomyopathy. Interactive CardioVascular and Thoracic Surgery. 2021; 33: 834–836.
[70]
Michelis KC, Zhong L, Tang WHW, Young JB, Peltz M, Drazner MH, et al. Durable Mechanical Circulatory Support in Patients with Amyloid Cardiomyopathy. Circulation: Heart Failure. 2020; 13: e007931.
[71]
DePasquale EC, Nasir K, Jacoby DL. Outcomes of adults with restrictive cardiomyopathy after heart transplantation. The Journal of Heart and Lung Transplantation. 2012; 31: 1269–1275.
[72]
Zangwill SD, Naftel D, L’Ecuyer T, Rosenthal D, Robinson B, Kirklin JK, et al. Outcomes of Children with Restrictive Cardiomyopathy Listed for Heart Transplant: a Multi-institutional Study. The Journal of Heart and Lung Transplantation. 2009; 28: 1335–1340.
[73]
Dipchand AI, Naftel DC, Feingold B, Spicer R, Yung D, Kaufman B, et al. Outcomes of Children with Cardiomyopathy Listed for Transplant: a Multi-institutional Study. The Journal of Heart and Lung Transplantation. 2009; 28: 1312–1321.
[74]
Maskatia SA, Decker JA, Spinner JA, Kim JJ, Price JF, Jefferies JL, et al. Restrictive Physiology is Associated with Poor Outcomes in Children with Hypertrophic Cardiomyopathy. Pediatric Cardiology. 2011; 33: 141–149.
[75]
Maron BJ. Risk stratification and role of implantable defibrillators for prevention of sudden death in patients with hypertrophic cardiomyopathy. Circulation Journal. 2010; 74: 2271–2282.
[76]
Chouairi F, Mullan CW, Sen S, Mori M, Fuery M, Elder RW, et al. Impact of the new heart allocation policy on patients with restrictive, hypertrophic, or congenital cardiomyopathies. PLoS ONE. 2021; 16: e0247789.
[77]
Towbin JA. Inherited cardiomyopathies. Circulation Journal. 2014; 78: 2347–2356.
[78]
Zaleta-Rivera K, Dainis A, Ribeiro AJS, Cordero P, Rubio G, Shang C, et al. Allele-Specific Silencing Ameliorates Restrictive Cardiomyopathy Attributable to a Human Myosin Regulatory Light Chain Mutation. Circulation. 2019; 140: 765–778.
[79]
Nguyen Q, Lim KRQ, Yokota T. Genome Editing for the Understanding and Treatment of Inherited Cardiomyopathies. International Journal of Molecular Sciences. 2020; 21: 733.
[80]
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, et al. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. International Journal of Molecular Sciences. 2019; 20: 4381.
[81]
Ma H, Marti-Gutierrez N, Park S, Wu J, Lee Y, Suzuki K, et al. Correction of a pathogenic gene mutation in human embryos. Nature. 2017; 548: 413–419.
[82]
Sen-Chowdhry S, Syrris P, McKenna WJ. Genetics of restrictive cardiomyopathy. Heart Failure Clinics. 2010; 6: 179–186.
[83]
Sipe JD, Benson MD, Buxbaum JN, Ikeda S, Merlini G, Saraiva MJM, et al. Amyloid fibril proteins and amyloidosis: chemical identification and clinical classification International Society of Amyloidosis 2016 Nomenclature Guidelines. Amyloid. 2016; 23: 209–213.
[84]
Conceição I, Damy T, Romero M, Galán L, Attarian S, Luigetti M, et al. Early diagnosis of ATTR amyloidosis through targeted follow-up of identified carriers of TTR gene mutations. Amyloid. 2019; 26: 3–9.
[85]
Anderson HN, Cetta F, Driscoll DJ, Olson TM, Ackerman MJ, Johnson JN. Idiopathic Restrictive Cardiomyopathy in Children and Young Adults. The American Journal of Cardiology. 2018; 121: 1266–1270.
[86]
Kawano H, Kawamura K, Kanda M, Ishijima M, Abe K, Hayashi T, et al. Histopathological changes of myocytes in restrictive cardiomyopathy. Medical Molecular Morphology. 2021; 54: 289–295.
[87]
Purevjav E, Arimura T, Augustin S, Huby A, Takagi K, Nunoda S, et al. Molecular basis for clinical heterogeneity in inherited cardiomyopathies due to myopalladin mutations. Human Molecular Genetics. 2012; 21: 2039–2053.
[88]
Callis TE, Jensen BC, Weck KE, Willis MS. Evolving molecular diagnostics for familial cardiomyopathies: at the heart of it all. Expert Review of Molecular Diagnostics. 2010; 10: 329–351.
[89]
Kaski JP, Syrris P, Burch M, Tomé-Esteban M, Fenton M, Christiansen M, et al. Idiopathic restrictive cardiomyopathy in children is caused by mutations in cardiac sarcomere protein genes. Heart. 2008; 94: 1478–1484.
[90]
Shah S, Yogasundaram H, Basu R, Wang F, Paterson DI, Alastalo TP, et al. Novel Dominant-Negative Mutation in Cardiac Troponin I Causes Severe Restrictive Cardiomyopathy. Circulation-Heart Failure. 2017; 10: e003820.
[91]
Duboscq-Bidot L, Xu P, Charron P, Neyroud N, Dilanian G, Millaire A, et al. Mutations in the Z-band protein myopalladin gene and idiopathic dilated cardiomyopathy. Cardiovascular Research. 2008; 77: 118–125.
[92]
Parvatiyar MS, Pinto JR, Dweck D, Potter JD. Cardiac troponin mutations and restrictive cardiomyopathy. Journal of Biomedicine & Biotechnology. 2010; 2010: 350706.
[93]
Arbustini E, Pasotti M, Pilotto A, Pellegrini C, Grasso M, Previtali S, et al. Desmin accumulation restrictive cardiomyopathy and atrioventricular block associated with desmin gene defects. European Journal of Heart Failure. 2006; 8: 477–483.
[94]
Hoedemaekers YM, Caliskan K, Majoor-Krakauer D, van de Laar I, Michels M, Witsenburg M, et al. Cardiac beta-myosin heavy chain defects in two families with non-compaction cardiomyopathy: linking non-compaction to hypertrophic, restrictive, and dilated cardiomyopathies. European Heart Journal. 2007; 28: 2732–2737.
[95]
Ware SM, Quinn ME, Ballard ET, Miller E, Uzark K, Spicer RL. Pediatric restrictive cardiomyopathy associated with a mutation in beta-myosin heavy chain. Clinical Genetics. 2008; 73: 165–170.
[96]
Meyer T, Ruppert V, Ackermann S, Richter A, Perrot A, Sperling SR, et al. Novel mutations in the sarcomeric protein myopalladin in patients with dilated cardiomyopathy. European Journal of Human Genetics. 2013; 21: 294–300.
[97]
Brodehl A, Ferrier RA, Hamilton SJ, Greenway SC, Brundler MA, Yu W, et al. Mutations in FLNC are Associated with Familial Restrictive Cardiomyopathy. Human Mutation. 2016; 37: 269–279..
[98]
Peled Y, Gramlich M, Yoskovitz G, Feinberg MS, Afek A, Polak-Charcon S, et al. Titin mutation in familial restrictive cardiomyopathy. International Journal of Cardiology. 2014; 171: 24–30.
[99]
Valdés-Mas R, Gutiérrez-Fernández A, Gómez J, Coto E, Astudillo A, Puente DA, et al. Mutations in filamin C cause a new form of familial hypertrophic cardiomyopathy. Nature Communications. 2014; 5: 5326.
[100]
Schuld J, Orfanos Z, Chevessier F, Eggers B, Heil L, Uszkoreit J, et al. Homozygous expression of the myofibrillar myopathy-associated p.W2710X filamin C variant reveals major pathomechanisms of sarcomeric lesion formation. Acta Neuropathologica Communications. 2020; 8: 154.
[101]
Dalkilic I, Schienda J, Thompson TG, Kunkel LM. Loss of FilaminC (FLNc) results in severe defects in myogenesis and myotube structure. Molecular and Cellular Biology. 2006; 26: 6522–6534.
[102]
Kimura K, Ooms A, Graf-Riesen K, Kuppusamy M, Unger A, Schuld J, et al. Overexpression of human BAG3P209L in mice causes restrictive cardiomyopathy. Nature Communications. 2021; 12: 3575.
[103]
Lewis AB. Clinical profile and outcome of restrictive cardiomyopathy in children. Am Heart J. 1992; 123: 1589–1593.
[104]
Webber SA. Primary restrictive cardiomyopathy in childhood. Prog Pediatr Cardiol. 2008; 25: 85–90.
[105]
Park J, Lee JM, Cho JS. Phenotypic Diversity of Cardiomyopathy Caused by an MYBPC3 Frameshift Mutation in a Korean Family: A Case Report. Medicina. 2021; 57: 281.
[106]
Nie L, Guo X, Esmailzadeh L, Zhang J, Asadi A, Collinge M, et al. Transmembrane protein ESDN promotes endothelial VEGF signaling and regulates angiogenesis. The Journal of Clinical Investigation. 2013; 123: 5082–5097.
[107]
Herrmann H, Cabet E, Chevalier NR, Moosmann J, Schultheis D, Haas J, et al. Dual Functional States of R406W-Desmin Assembly Complexes Cause Cardiomyopathy With Severe Intercalated Disc Derangement in Humans and in Knock-In Mice. Circulation. 2020; 142: 2155–2171.
[108]
Tucker NR, McLellan MA, Hu D, Ye J, Parsons VA, Mills RW, et al. Novel Mutation in FLNC (Filamin C) Causes Familial Restrictive Cardiomyopathy. Circulation: Cardiovascular Genetics. 2017; 10: e001780.
[109]
Kiselev A, Vaz R, Knyazeva A, Khudiakov A, Tarnovskaya S, Liu J, et al. De novo mutations in FLNC leading to early-onset restrictive cardiomyopathy and congenital myopathy. Human Mutation. 2018; 39: 1161–1172.
[110]
Schubert J, Tariq M, Geddes G, Kindel S, Miller EM, Ware SM. Novel pathogenic variants in filamin C identified in pediatric restrictive cardiomyopathy. Human Mutation. 2018; 39: 2083-2096.
[111]
Hinton RB, Michelfelder EC, Marino BS, Bove KE, Ware SM. A fetus with hypertrophic cardiomyopathy, restrictive, and single-ventricle physiology, and a beta-myosin heavy chain mutation. Journal of Pediatrics. 2010; 157: 164–166.
[112]
Karam S, Raboisson M, Ducreux C, Chalabreysse L, Millat G, Bozio A, et al. A de novo mutation of the beta cardiac myosin heavy chain gene in an infantile restrictive cardiomyopathy. Congenital Heart Disease. 2008; 3: 138–143.
[113]
Andersen PS, Havndrup O, Bundgaard H, Moolman-Smook JC, Larsen LA, Mogensen J, et al. Myosin light chain mutations in familial hypertrophic cardiomyopathy: phenotypic presentation and frequency in Danish and South African populations. Journal of Medical Genetics. 2001; 38: E43.
[114]
Olson TM, Karst ML, Whitby FG, Driscoll DJ. Myosin light chain mutation causes autosomal recessive cardiomyopathy with mid-cavitary hypertrophy and restrictive physiology. Circulation. 2002; 105: 2337–2340.
[115]
Yuan C, Kazmierczak K, Liang J, Kanashiro-Takeuchi R, Irving TC, Gomes AV, et al. Hypercontractile mutant of ventricular myosin essential light chain leads to disruption of sarcomeric structure and function and results in restrictive cardiomyopathy in mice. Cardiovascular Research. 2017; 113: 1124–1136.
[116]
Huby A, Mendsaikhan U, Takagi K, Martherus R, Wansapura J, Gong N, et al. Disturbance in Z-Disk Mechanosensitive Proteins Induced by a Persistent Mutant Myopalladin Causes Familial Restrictive Cardiomyopathy. Journal of the American College of Cardiology. 2014; 64: 2765–2776.
[117]
Mogensen J, Kubo T, Duque M, Uribe W, Shaw A, Murphy R, et al. Idiopathic restrictive cardiomyopathy is part of the clinical expression of cardiac troponin i mutations. The Journal of Clinical Investigation. 2003; 111: 209–216.
[118]
Wen Y, Xu Y, Wang Y, Pinto JR, Potter JD, Kerrick WGL. Functional Effects of a Restrictive-Cardiomyopathy-Linked Cardiac Troponin i Mutation (R145W) in Transgenic Mice. Journal of Molecular Biology. 2009; 392: 1158–1167.
[119]
Lindert S, Cheng Y, Kekenes-Huskey P, Regnier M, McCammon JA. Effects of HCM cTnI mutation R145G on troponin structure and modulation by PKA phosphorylation elucidated by molecular dynamics simulations. Biophysical Journal. 2015; 108: 395–407.
[120]
Yumoto F, Lu Q, Morimoto S, Tanaka H, Kono N, Nagata K, et al. Drastic Ca2+ sensitization of myofilament associated with a small structural change in troponin i in inherited restrictive cardiomyopathy. Biochemical and Biophysical Research Communications. 2005; 338: 1519–1526.
[121]
Jean-Charles P, Li Y, Nan C, Huang X. Insights into restrictive cardiomyopathy from clinical and animal studies. Journal of Geriatric Cardiology. 2011; 8: 168–183.
[122]
Gomes AV, Liang J, Potter JD. Mutations in human cardiac troponin i that are associated with restrictive cardiomyopathy affect basal ATPase activity and the calcium sensitivity of force development. The Journal of Biological Chemistry. 2005; 280: 30909–30915.
[123]
Davis J, Wen H, Edwards T, Metzger JM. Thin filament disinhibition by restrictive cardiomyopathy mutant R193H troponin i induces Ca2+-independent mechanical tone and acute myocyte remodeling. Circulation Research. 2007; 100: 1494–1502.
[124]
Peddy SB, Vricella LA, Crosson JE, Oswald GL, Cohn RD, Cameron DE, et al. Infantile restrictive cardiomyopathy resulting from a mutation in the cardiac troponin T gene. Pediatrics. 2006; 117: 1830–1833.
[125]
Menon SC, Michels VV, Pellikka PA, Ballew JD, Karst ML, Herron KJ, et al. Cardiac troponin T mutation in familial cardiomyopathy with variable remodeling and restrictive physiology. Clinical Genetics. 2008; 74: 445–454.
[126]
Miller T, Szczesna D, Housmans PR, Zhao J, de Freitas F, Gomes AV, et al. Abnormal contractile function in transgenic mice expressing a familial hypertrophic cardiomyopathy-linked troponin T (i79N) mutation. The Journal of Biological Chemistry. 2001; 276: 3743–3755.
[127]
Hoshijima M. Mechanical stress-strain sensors embedded in cardiac cytoskeleton: Z disk, titin, and associated structures. American Journal of Physiology-Heart and Circulatory Physiology. 2006; 290: H1313–H1325.
[128]
Cimiotti D, Budde H, Hassoun R, Jaquet K. Genetic Restrictive Cardiomyopathy: Causes and Consequences-An Integrative Approach. International Journal of Molecular Sciences, 2021; 22: 558.
[129]
Solaro RJ, Kobayashi T. Protein phosphorylation and signal transduction in cardiac thin filaments. The Journal of Biological Chemistry. 2011; 286: 9935–9940.
[130]
Chen S, Balfour IC, Jureidini S. Clinical spectrum of restrictive cardiomyopathy in children. J Heart Lung Transplant. 2001; 20: 90–92.
[131]
Purevjav E. Animal Models of Cardiomyopathies. IntechOpen: United Kingdom. 2020.
[132]
Mosqueira D, Mannhardt I, Bhagwan JR, Lis-Slimak K, Katili P, Scott E, et al. CRISPR/Cas9 editing in human pluripotent stem cell-cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy. European Heart Journal. 2018; 39: 3879–3892.
[133]
Johansen AK, Molenaar B, Versteeg D, Leitoguinho AR, Demkes C, Spanjaard B, et al. Postnatal Cardiac Gene Editing Using CRISPR/Cas9 with AAV9-Mediated Delivery of Short Guide RNAs Results in Mosaic Gene Disruption. Circulation Research. 2017; 121: 1168–1181.
[134]
Rai TS, Ahmad S, Bahl A, Ahuja M, Ahluwalia TS, Singh B, et al. Genotype phenotype correlations of cardiac beta-myosin heavy chain mutations in Indian patients with hypertrophic and dilated cardiomyopathy. Molecular and Cellular Biochemistry. 2009; 321: 189–196.
[135]
Kubo T, Gimeno JR, Bahl A, Steffensen U, Steffensen M, Osman E, et al. Prevalence, clinical significance, and genetic basis of hypertrophic cardiomyopathy with restrictive phenotype. Journal of the American College of Cardiology. 2007; 49: 2419–2426.
[136]
Kimura A, Harada H, Park JE, Nishi H, Satoh M, Takahashi M, et al. Mutations in the cardiac troponin I gene associated with hypertrophic cardiomyopathy. Nature Genetics. 1997; 16: 379–382.
[137]
Mogensen J, Murphy RT, Kubo T, Bahl A, Moon JC, Klausen IC, et al. Frequency and clinical expression of cardiac troponin i mutations in 748 consecutive families with hypertrophic cardiomyopathy. Journal of the American College of Cardiology. 2004; 44: 2315–2325.
[138]
Tsuru H, Ishida H, Narita J, Ishii R, Suginobe H, Ishii Y, et al. Cardiac Fibroblasts Play Pathogenic Roles in Idiopathic Restrictive Cardiomyopathy. Circulation Journal. 2021; 85: 677–686.
[139]
Kimura Y, Karakama S, Hirakawa A, Tsuchiaka S, Kobayashi M, Machida N. Pathological Features and Pathogenesis of the Endomyocardial Form of Restrictive Cardiomyopathy in Cats. Journal of Comparative Pathology. 2016; 155: 190–198.
[140]
Sehnert AJ, Huq A, Weinstein BM, Walker C, Fishman M, Stainier DYR. Cardiac troponin T is essential in sarcomere assembly and cardiac contractility. Nature Genetics. 2002; 31: 106–110.
[141]
Mogensen J, Hey T, Lambrecht S. A Systematic Review of Phenotypic Features Associated With Cardiac Troponin I Mutations in Hereditary Cardiomyopathies. Canadian Journal of Cardiology. 2015; 31: 1377–1385.
[142]
Liu X, Zhang L, Pacciulli D, Zhao J, Nan C, Shen W, et al. Restrictive Cardiomyopathy Caused by Troponin Mutations: Application of Disease Animal Models in Translational Studies. Frontiers in Physiology. 2016; 7: 629.
[143]
Jean-Charles P, Du J, Li Y, Gobara N, Huang X. Abstract 843: Restrictive Cardiomyopathy Linked cTnI Mutation (K178E) Causes Severe Heart Failure and Early Mortality. Circulation. 2008; 118: S_623.
[144]
Zhao W, Qian lu, Luo J, Pan B, Liu L, Tian J. Cardiac troponin i R193H mutant interacts with HDAC1 to repress phosphodiesterase 4D expression in cardiomyocytes. Genes & Diseases. 2021; 8: 569–579.
[145]
Ueno M, Takeda A, Yamazawa H, Takei K, Furukawa T, Suzuki Y, et al. A case report: Twin sisters with restrictive cardiomyopathy associated with rare mutations in the cardiac troponin i gene. Journal of Cardiology Cases. 2020; 23: 154–157.
[146]
Dvornikov AV, Smolin N, Zhang M, Martin JL, Robia SL, de Tombe PP. Restrictive Cardiomyopathy Troponin i R145W Mutation does not Perturb Myofilament Length-dependent Activation in Human Cardiac Sarcomeres. Journal of Biological Chemistry. 2016; 291: 21817–21828.
[147]
Madan A, Viswanathan MC, Woulfe KC, Schmidt W, Sidor A, Liu T, et al. TNNT2mutations in the tropomyosin binding region of TNT1 disrupt its role in contractile inhibition and stimulate cardiac dysfunction. Proceedings of the National Academy of Sciences. 2020; 117: 18822–18831.
[148]
Yadav S, Sitbon YH, Kazmierczak K, Szczesna-Cordary D. Hereditary heart disease: pathophysiology, clinical presentation, and animal models of HCM, RCM, and DCM associated with mutations in cardiac myosin light chains. Pflugers Arch - Eur J Physiol. 2019; 471: 683–699.
[149]
Caleshu C, Sakhuja R, Nussbaum RL, Schiller NB, Ursell PC, Eng C, et al. Furthering the link between the sarcomere and primary cardiomyopathies: restrictive cardiomyopathy associated with multiple mutations in genes previously associated with hypertrophic or dilated cardiomyopathy. American Journal of Medical Genetics, Part A. 2011; 155A: 2229–2235.
[150]
Tharp CA, Haywood ME, Sbaizero O, Taylor MRG, Mestroni L. The Giant Protein Titin’s Role in Cardiomyopathy: Genetic, Transcriptional, and Post-translational Modifications of TTN and Their Contribution to Cardiac Disease. Frontiers in Physiology. 2019; 10: 1436.
[151]
Eden M, Frey N. Cardiac Filaminopathies: Illuminating the Divergent Role of Filamin C Mutations in Human Cardiomyopathy. Journal of Clinical Medicine. 2021; 10: 577.
[152]
Ader F, De Groote P, Réant P, Rooryck-Thambo C, Dupin-Deguine D, Rambaud C, et al. FLNC pathogenic variants in patients with cardiomyopathies: Prevalence and genotype-phenotype correlations. Clinical Genetics. 2019; 96: 317–329.
[153]
Ho CY, Jaalouk DE, Vartiainen MK, Lammerding J. Lamin a/C and emerin regulate MKL1–SRF activity by modulating actin dynamics. Nature. 2013; 497: 507–511.
[154]
Sullivan T, Escalante-Alcalde D, Bhatt H, Anver M, Bhat N, Nagashima K, et al. Loss of a-type lamin expression compromises nuclear envelope integrity leading to muscular dystrophy. The Journal of Cell Biology. 1999; 147: 913–920.
[155]
Paller MS, Martin CM, Pierpont ME. Restrictive cardiomyopathy: an unusual phenotype of a lamin a variant. ESC Heart Failure. 2018; 5: 724–726.
[156]
Palmer BM, McConnell BK, Li GH, Seidman CE, Seidman JG, Irving TC, et al. Reduced cross-bridge dependent stiffness of skinned myocardium from mice lacking cardiac myosin binding protein-C. Molecular and Cellular Biochemistry. 2004; 263: 73–80.
[157]
Bang ML, Mudry RE, McElhinny AS, Trombitás K, Geach AJ, Yamasaki R, et al. Myopalladin, a novel 145-kilodalton sarcomeric protein with multiple roles in Z-disc and i-band protein assemblies. The Journal of Cell Biology. 2001; 153: 413–427.
[158]
Kojic S, Nestorovic A, Rakicevic L, Belgrano A, Stankovic M, Divac A, et al. A novel role for cardiac ankyrin repeat protein Ankrd1/CARP as a co-activator of the p53 tumor suppressor protein. Archives of Biochemistry and Biophysics. 2010; 502: 60–67.
[159]
Mendsaikhan U, Martherus R, Takagi K, Huby AC, Gong N, Kramer K, et al. Abstract 13400: Myopalladin Mutation Induced ERK1/2 and CARP/ANKRD1 Dysregulation in Restrictive Cardiomyopathy. Circulation. 2013; 128: A13400.
[160]
Gu Q, Mendsaikhan U, Khuchua Z, Jones BC, Lu L, Towbin JA, et al. Dissection of Z-disc myopalladin gene network involved in the development of restrictive cardiomyopathy using system genetics approach. World Journal of Cardiology. 2017; 9: 320–331.
[161]
Stürner E, Behl C. The Role of the Multifunctional BAG3 Protein in Cellular Protein Quality Control and in Disease. Frontiers in Molecular Neuroscience. 2017; 10: 177.
[162]
Fang X, Bogomolovas J, Zhou PS, Mu Y, Ma X, Chen Z, et al. P209L mutation in Bag3 does not cause cardiomyopathy in mice. American Journal of Physiology-Heart and Circulatory Physiology. 2019; 316: H392–H399.
[163]
Quintana MT, Parry TL, He J, Yates CC, Sidorova TN, Murray KT, et al. Cardiomyocyte-Specific Human Bcl2-Associated Anthanogene 3 P209L Expression Induces Mitochondrial Fragmentation, Bcl2-Associated Anthanogene 3 Haploinsufficiency, and Activates p38 Signaling. The American Journal of Pathology. 2016; 186: 1989–2007.
[164]
Brodehl A, Gaertner-Rommel A, Milting H. Molecular insights into cardiomyopathies associated with desmin (DES) mutations. Biophysical Reviews. 2018; 10: 983–1006.
[165]
Walsh MA, Grenier MA, Jefferies JL, Towbin JA, Lorts A, Czosek RJ. Conduction abnormalities in pediatric patients with restrictive cardiomyopathy. Circulation. Heart Failure. 2012; 5: 267–273.
[166]
Alhamoudi KM, Barhoumi T, Al-Eidi H, Asiri A, Nashabat M, Alaamery M, et al. A homozygous nonsense mutation in DCBLD2 is a candidate cause of developmental delay, dysmorphic features and restrictive cardiomyopathy. Scientific Reports. 2021; 11: 12861.
[167]
Sato T, Ito M. Fibroblast-Cardiomyocyte Interaction in Pediatric Restrictive Cardiomyopathy. Circulation Journal. 2021; 85: 687–689.
Share
Back to top