Abstract
Experimental models of cardiac disease play a key role in understanding the pathophysiology of the disease and developing new therapies. The features of the experimental models should reflect the clinical phenotype, which can have a wide spectrum of underlying mechanisms. We review characteristics of commonly used experimental models of cardiac physiology and pathophysiology in all translational steps including in vitro, small animal, and large animal models. Understanding their characteristics and relevance to clinical disease is the key for successful translation to effective therapies.
Keywords: primary cardiomyocytes, neonatal cardiomyocytes, ischemic, non-ischemic, pressure overload, volume overload
1. Introduction
Heart diseases represented by coronary artery disease and heart failure are increasing worldwide(1). Researchers devote intense efforts to prevent and cure heart diseases in both clinical and experimental research areas. Experimental research has played and will continue to play key roles in discovering disease mechanisms and developing new therapies. In this review, we provide characteristics of commonly used experimental models of cardiac physiology and pathophysiology by covering all translational steps from in vitro to large animal models (Table 1)
Table 1.
Comparisons of experimental models
In vitro | Ex vivo | In vivo | |
---|---|---|---|
Features |
|
|
|
2. In Vitro models
First, we discuss in vitro models of cardiac disease. Although various types of cells compose the heart, cardiomyocytes account for a majority of heart mass and predominantly affect cardiac function(2,3). Since malfunction of cardiomyocytes is a causative mechanism in most heart diseases(4), a large amount of effort was spent to develop multiple in vitro cardiomyocyte models. The in vitro model system offers more precise control of experimental conditions and manipulations, which provide many advantages over the in vivo models in signaling pathway studies or high-throughput drug screenings (Table 2). For more than three decades, several cell culture tools, including primary cells, immortalized cell lines, human embryonic- and induced pluripotent stem cell (ESC and iPSC, respectively) derived cardiomyocytes have been developed to study heart disease in various settings. Herein, we discuss different types of cells used to model heart disease and their essential characteristics.
Table 2.
Comparisons of in vitro models
Neonatal cardiomyocytes | Adult cardiomoycytes | Cardiac cell lines | ESC/iPSCs | |
---|---|---|---|---|
Pros |
|
|
|
|
Cons |
|
|
|
|
Application |
|
|
|
|
2-1. Neonatal cardiomyocytes
Rodent neonatal cardiomyocytes are the most widely used in vitro models in cardiac research. Since the neonatal cardiomyocytes are isolated from 1~5-days old rats/mice, the cells are yet immature in morphology and transcriptional profiles(5–7). Despite this limitation, neonatal cardiomyocytes have been the workhorse of cardiac research, because of their relatively easy isolation, gene expression manipulation capabilities, and reliable physiologic microenvironment(8–10). Harary and Farley(11) first developed techniques for isolation of neonatal cardiomyocytes more than 50 years ago. Fundamental procedures to isolate and culture the cells have been optimized by many researchers over the years. Thanks to these efforts, commercially available kits now offer easy isolation of neonatal cardiomyocytes for researchers without extensive experiences. Rats are commonly used for their advantages over other species, including low cost, higher yields of viable cells and efficient transfection rates for gene manipulation(8–10). Meanwhile, because of a large number of transgenic lines, mice are also employed to study functional roles of specific genes and proteins(12).
Neonatal rat cardiomyocyte (NRCM) model offers great advantages for studying cardiac hypertrophy. In 1982, Simpson et al.(13) demonstrated induction of cardiac hypertrophy in NRCM culture by norepinephrine treatment. This study opened the door for using this model as a platform for studying hypertrophy in vitro. Since then, numerous compounds and growth factors have been tested in NRCM culture and number of molecules have been identified to induce pathological hypertrophy, including phenylephrine(14–16), angiotensin II(17,18), endothelin-1(17,19), and the diacylglycerol mimetic phorbol 12-myristate 12-acetate(20). In response to these stimulations, NRCMs increase both volume and cell surface area, promote sarcomeric organization and reactivate fetal gene expression(13,16,20). Therefore, assessment of these profiles is central to evaluate cardiac hypertrophy in the NRCM model. Recent advances in fluorescence-based staining methods and image-analyzing software enable accurate measurements of cell surface area(17). Staining with phalloidin-Rhodamine, cardiac troponin I or α-actinin is a widely-accepted method to assess sarcomeric organization(21,22). Reactivations of a fetal gene program, such as atrial natriuretic factor, brain natriuretic peptide, α-skeletal muscle actin, and β-myosin heavy chain can be evaluated by qRT-PCR(14).
Sustained increase in mechanical cardiac load is an important pathological factor that promotes cardiac diseases. The sustained stretch model using NRCMs enables in vitro replication of increased mechanical load in isolated cells. Komuro et al.(23,24) developed deformable culture dishes using silicone membrane to simulate the stretch-induced stress in 1990. Currently, several apparatuses have been developed to provide computer-controlled sustained or cyclic stretch. Sustained stretch induces dynamic changes in gene expression in time and stretch-dependent manner. These changes subsequently induce apoptosis, autophagy and hypertrophy in NRCMs(25–29). Fluorescence-based staining to detect hypertrophic, apoptotic or autophagic markers and evaluation of gene expression profiles related to these signaling is usually employed to determine stretch-induced pathology(25–29).
The NRCM model is also a useful tool to study ischemia/reperfusion (I/R) injury. In 1977, Acosta and Puckett first reported effects of hypoxic environment on in vitro cell culture(30). Nowadays different levels of hypoxic environments can be generated by culturing cells in hypoxic chambers with different concentrations of gas mixtures, including anoxic condition (e.g. 5 % CO2 and 95 % N2)(31), and also by the treatment with oxygen-scavenging compounds (e.g., Na2S2O4) (32). Accumulation of reactive oxygen species (ROS), lipid peroxidation, increased membrane permeability and apoptosis represent the impacts of I/R injury(33–36). Staining of ROS by chemical indicator (e.g., MitoSOX)(31), measurement of SOD2 and catalase enzymatic activities(31), flow cytometry-based apoptosis detection(37,38) and western blotting with apoptotic markers (e.g., caspase 3, caspase 7, Bcl-2, Bcl-xl)(35) are used for evaluating impact of I/R injury.
Advantages of neonatal cardiomyocytes over adult cardiomyocytes include relatively ease of culture and high transfection efficiency with liposomal transfection methods. Nevertheless, they have some limitations. Most notably, neonatal cardiomyocytes lack a definitive t-tubular subsystem(5). Some components of signaling pathways differ between neonatal and adult myocytes(39), implying less accurate reflection of Ca2+ dynamics and signaling compared to adult cardiomyocytes. Non-uniform cell shortening is another limitation for modeling of contractile measurements in these cells. Hence, neonatal cardiomyocytes are most powerful in quickly and easily determining the effects of the gene manipulation and screening lead compounds through high-throughput drug screening assays(40,41). Ideally, the results obtained from neonatal cardiomyocytes should be validated with similarly designed experiments using adult cardiomyocytes.
2-2. Adult cardiomyocytes
Adult cardiomyocytes best recapitulate the morphology and behavior of the cells in intact human heart. These cells are rod-shaped, binucleated, and have well-organized sarcomeres throughout the cell body. In 1895, Oscar Langendorff first developed a protocol to isolate adult cardiomyocytes by the retrograde perfusion of the heart with an enzymatic solution and current researchers follow protocols that are similar to the original in principle(42). The yield and quality of the isolated cardiomyocytes are affected considerably by the heart cannulation and perfusion. Despite the technical difficulty, however, their above and below mentioned advantages make adult cardiomyocytes as one of the most frequently-used models for in vitro research.
The key advantage of adult cardiomyocytes for in vitro studies may be the wide-applicability of the isolation protocol to various types of animals, in contrast to the neonatal cardiomyocytes. The animals of all ages can be used to study the effects of aging(43). Adult cardiomyocytes isolated from male versus females allow probing the effects of sex difference on cardiac function, which is not possible with neonatal cardiomyocytes or immortalized cell lines(44). Furthermore, animals with diverse disease conditions can be utilized for this model. For example, the heart after surgical disease induction or cardio-toxic chemicals can be used to isolate cells, and studied for their structural and functional properties at cellular levels(45–48). A large number of genetic models also make this model attractive. Indeed, isolated cardiomyocytes from transgenic mice were utilized to characterize the roles of genetic mutations found in familial dilated cardiomyopathy patients(49–51).
Another advantage of adult cardiomyocyte is a wide-spectrum of applicable experimental assays. Since adult cardiomyocytes have a mature sarcomeric structure and ion channels, different methods including patch-clamp(52), contractility measurements(53), and Ca2+ imaging studies(54,55) can be applied. These methods provide in depth analysis of contractility, sarcoplasmic reticulum Ca2+ load, diastolic Ca2+ levels and myofilament Ca2+ sensitivity. For example, Kerr et al.(56) showed disruption of Ca2+ signaling using several methods such as measurements of contractility, Ca2+ transient, stretch-induced ROS production, stretch-induced Ca2+ influx and Ca2+ sparks in isolated cardiomyocytes from a mouse model of Duchenne muscular dystrophy (MDX mice). Through this study, the authors elucidated how post-translational modification of α-tubulin affects Ca2+ signaling that underlies Duchenne muscular dystrophy pathologies.
Adult cardiomyocytes are also a useful model to study cardiac hypertrophy. Hypertrophy is induced in adult cardiomyocytes in response to many different triggers, including norepinephrine(57), phenylephrine(58), angiotensin II(59) and isoproterenol(60). Similar to neonatal cardiomyocytes, these stimulations activate fetal gene program and increase the abundance of myosin heavy chain protein, the rate of protein synthesis and total protein levels. Neonatal cardiomyocytes show dramatic changes in cell size (~150%) and sarcomeric organization within 48h of treatment of the triggers(13). However, this is not always the case in adult cardiomyocytes, and the shape of cardiomyocytes is closely related to the contractile function and cell condition. Therefore, instead of the image-based morphometric analysis, qRTPCR and western blotting of fetal genes are widely-accepted methods to detect hypertrophic features in adult cardiomyocytes. [3H] phenylalanine incorporation and colorimetric protein quantitation (e.g., Bradford assay, bicinchoninic acid assay) are also employed to verify the changes in the rate of protein synthesis and total protein levels by hypertrophic stimulations(58,61).
Similar to the neonatal cardiomyocytes, adult cardiomyocytes are also able to be utilized for mimicking mechanical overload or I/R injury by the sustained stretch or hypoxic environments. These stresses turn on the similar signaling pathways with neonatal cardiomyocytes including hypertrophic, apoptotic and autophagic responses, but also induce sarcoplasmic reticulum Ca2+ leak and cardiac dysfunction(62,63). Overall, adult cardiomyocytes offer very reliable and efficient models in studying cardiac pathologies.
2-3. Immortalized cell lines
As discussed above, primary cells (adult cardiomyocytes, neonatal cardiomyocytes) are the most accepted in vitro models for studying cardiac diseases. However, these cells have some practical problems that make them challenging to use. Since the majority of cardiomyocytes differentiate terminally in vivo in the perinatal period, they cannot be passaged in culture. These cells are fragile and difficult to maintain in culture for long periods. Especially, cultures of primary cardiomyocytes maintained for several weeks undergo morphological and functional changes over time and yield a heterogeneous population of cells(64,65). The difficulties to transfect and recover from frozen stocks are also considerable issues in these cells. In addition, animal sacrifice is necessary for the use of primary cells, which leads to practical and ethical limitations. To address these issues, many attempts have been made to establish immortalized cardiac cell lines. Various groups have developed suitable cell culture systems including H9C2(66), ANT-T-antigen(67), AT-1cells(68), MC29(69), HL-1(70), and AC16(71). Since immortalized cell lines retain similar gene expression profiles and phenotypic features of the cardiomyocytes, these cell lines have commonly served as an alternative to primary cells.
2-3a. H9C2
The H9c2 cells, myoblast cell line, were first isolated from embryonic BDIX rat ventricular tissue in 1976(66). At this stage, cells were not sufficiently differentiated into adult cardiomyocytes yet and lacked a clear cardiac phenotype. However, Menard et al.(72) established a protocol to differentiate them from mono-nucleated myoblasts to cardiac-like myotubes by the addition of all-trans retinoic acid (RA) to a 1% serum media. According to previous total genome microarray study, transcriptome alterations during H9c2 cell differentiation promote the cells to express cardiac-specific proteins, including cardiac troponins, calsequestrin, ryanodine receptor and sarco (endo) plasmic reticulum calcium ATPase(73). Therefore, H9c2 cells need to undergo the differentiation procedure for proper use of this cell. H9c2 cells lack the ability to contract but shows many similarities to primary cardiomyocytes, and thus have been used for investigating the molecular and cellular processes involved in cardiac hypertrophy(74), apoptosis(75), I/R injury(76), and toxicology(77). Notably, in the parallel comparison with NRCMs, H9c2 cells showed a more robust increase in cell surface area and similar levels of fetal gene activation to NRCM(74). Moreover, in another comparison study, H9c2 cells showed more similarity to primary cardiomyocytes than HL-1 cells with regard to energy metabolism patterns, such as cellular ATP levels, bioenergetics, metabolism, function and morphology of mitochondria(76). Based on these data, H9c2 cells are widely accepted as a valid in vitro system to study cardiac disease.
2-3b. HL-1
The HL-1 cell line originated from AT-1 cardiac myocytes. AT-1 cells are immortalized atrial cardiomyocytes derived from an atrial tumor growing transgenic mice with the simian virus 40 (SV40) large T antigen expression(68). Although AT-1 cells maintained a cardiomyocyte phenotype and ability to contract, they could not be serially passaged and revived from cryopreserved stocks. In 1998, Claycomb et al.(70) established the HL-1cell line to improve the limitations of AT-1 cells. The author reported that HL-1 cells maintain cardiac morphological, biochemical, and electrophysiological properties and solves the problems in AT-1 cells. HL-1 cells express numerous signaling receptors expressed in adult cardiomyocytes, and respond similarly to their agonists in pharmacological studies, which suggest that HL-1 cells exhibit an adult cardiomyocyte-like gene expression profile(70,78,79). Indeed, in the proteomic analysis of HL-1 cells in comparison with patient cardiac samples, HL-1 cells exerted similar alterations in the proteome to the biopsy samples taken from patients with ischemic cardiomyopathy, including cell death pathways and oxidative stress response(80). Another important feature of HL-1 cells is the ability to spontaneously contract. HL-1 cells contain highly organized sarcomeres and express the necessary ion channels required for generating action potentials characteristic of primary cardiomyocytes(70,81). Sartiani et al.(82) reported that the electrophysiological properties of HL-1 cells are sufficiently similar to those of primary cardiomyocytes for testing pharmacological drugs acting on ion currents. Meanwhile, HL-1 cells need to be maintained in medium containing adenosine, retinoic acid, and norepinephrine to maintain a differentiated phenotype and stimulate beating(70). These requirements may limit their usefulness in developing models of cardiac disease. Moreover, atrial cell-like gene profiles somewhat limit their utility for studying ventricular failure(83). Nevertheless, HL-1 cells have been used in various in vitro settings(76,84–86), and are an efficient alternative for primary cells.
2-3c. AC16
AC16 cells are the cell line that is established most recently. Since this cell line is derived from human ventricular tissues by fusion with SV40 transformed human fibroblasts, it is expected to recapitulate human cardiomyocytes more similarly than other cell types. Dividson et al.(71) reported that AC16 cells express cardiomyocyte-specific markers, such as transcription factors, contractile proteins, muscle-specific intermediate filament protein and the cardiomyocyte-specific peptide hormones. Moreover, these cells appear to express functional gap junctions and myofibrils, which indicate high similarity to adult cardiomyocytes. AC16 cells have been tested in many in vitro disease studies, including cardiac hypertrophy, I/R injury, oxidative stress and toxicology studies(87–91). Although many studies support the usefulness of AC16 cells as an in vitro model system for cardiac research, no study has compared AC16 cells to primary cells yet. Thus, one should use with caution when interpreting data obtained from AC16 cells.
2-4. ESC and iPSC derived cardiomyocytes
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the capacity to self-renew by dividing and pluripotency to differentiate to all cell types. While ESCs are derived from different sources of blastocysts that are already pluripotent(92–94), iPSCs are generated from somatic cells by the introduction of defined transcription factors, such as OCT4, SOX1, KLF4, and c-MYC(95). ESC or iPSC-derived cardiomyocytes are the cells that are induced to differentiate to cardiomyocytes from these pluripotent stem cells. The research using these stem cells has grown dramatically in recent years to develop an efficient in vitro assay using human cells for personalized medicine. For example, stem cell-derived cardiomyocytes enable prediction of cardiotoxicity in respective patient to increase the safety of novel drugs. Peters et al.(96) introduced a novel assay to predict embryotoxicity by the automatic record of contraction of ESC-derived cardiomyocytes. Recently, this assay system was applied for screening embryotoxicity of commonly-used compounds in food and cosmetic production(97). Patients with certain genetic backgrounds exhibit increased cardiotoxic sensitivity to treatment with doxorubicin, and this susceptibility was recapitulated in iPSC-derived cardiomyocyte cultures(98,99). However, these stem cell-based models are not well suited for cardiac physiology applications at the moment. Compared to other model systems, the cultures of ESCs or iPSCs are technically challenging and cost ineffective. The efficiency of proliferation and differentiation remains low and long-term culture as well as several supplements to induce differentiation makes this system technically difficult and expensive. Moreover, although the spontaneously beating ESC or iPSC-derived cardiomyocytes expressed early cardiomyocyte lineage markers, they remained phenotypically immature and were more similar to neonatal cardiomyocytes than adult cardiomyocytes(100–102). In addition, stem cell-derived cardiomyocytes are not able to be maintained as a cell line and hence they are dependent on new isolations, which cause batch-to-batch-variations. For these reasons, the stem cell-based models have not yet replaced animal-based models in cardiac physiology research. Nevertheless, their ability to study patient unique genetic disorder on dish is a significant advantage. ESC and iPSC can be derived from patients suffering specific genetic diseases, and differentiated into cardiomyocytes that retain those disease-specific traits. Unlike other models, genetic manipulations are not required and even the causative mutation does not have to be known for generating a model. For example, a number of genetic mutations behind hypertrophic cardiomyopathy, the most common inheritable heart disease, have been identified using these models. Carvajal-Vergara et al.(103) generated iPSC from patients with LEOPARD syndrome (Lentigines, Electrocardiographic abnormalities, Ocular hypertelorism, Pulmonary valve stenosis, Abnormal genitalia, Retardation of growth and Deafness), a kind of HCM, that have a mutation at Thr468 in the PTPN11 gene. The cells retained the phenotype in patients with LEOPARD syndrome, such as increase in cell size, sarcomeric organization and preferential localization of NFATc4 in the nucleus. Using these cells, the authors discovered RAS-MAPK signal transduction as a novel signaling pathways that promote the disease phenotype. Besides, the stem cell-based model has been applied to study various genetic disorders, including familial dilated cardiomyopathy(104), Duchenne muscular dystrophy(105), Friedreich’s ataxia(106) and catecholaminergic polymorphic ventricular tachycardia(107). In summary, stem cell-based models are promising, but some improvements are yet necessary in the proliferation, differentiation, and maturation of the cells. Current use mostly focuses on personalized screening of the drugs and studies on specific gene mutations.
Supplementation of functional cardiomyocytes for dysfunctional hearts by trans-differentiated ESC or iPSC is an attractive therapeutic option. As of 2018, 29 clinical trials involving hESC-derived stem cell products and three trials that involve hiPSC derivatives have been approved(108). Nevertheless, there seem to be several hurdles for these approaches to become a daily clinical practice. Common to both ESC and iPSC are potential risks of teratoma originating from residual pluripotent cells, the occurrence of arrhythmias and alloimmunization. Moreover, the efficiency of cardiomyogenic differentiation remains a challenge for both cell types. The key advantage of iPSC is the potential autologous application, whereas ESC lines are better standardized at the moment. Understanding the detailed characteristics as well as fine-tuning the trans-differentiation of ESC and iPSC cardiomyocytes is of paramount importance for therapeutic application.
2-5. Engineered cardiac tissues
While individual cell models provide important insights in cardiomyocytes pathophysiology, they do not account for cell-to-cell interactions, particularly those through direct contacts. In vitro tissue culture models incorporate multi-cellular component and three-dimensional structure of the heart muscle in the model system, allowing assessment of more complex interactions between the cells in cardiac tissue. Engineered cardiac tissues are produced by culturing cardiomyocytes together with other cell types. These cultured engineered tissues are attached to microsensors that measure forces generated by the tissue. Effects of drugs or biological substances on cardiac tissue contraction can be evaluated using this system(109,110), and tissue-on-chip tools are developed to enable high-throughput drug screening using engineered cardiac tissues(111). Efforts to create engineered tissues with pumping function are also being made(112,113).
3. Ex Vivo models
Assessment of the heart function as a pump system cannot be examined in in vitro system at the moment. Explanted heart allows detailed characterizations of cardiac pump function and electrophysiology in a well-controlled manner. Langendorff perfusion is the most common method employed and uses retrograde perfusion of oxygenate physiological buffer(114). Hearts from various species have been studied using this approach including that from humans(115). Pressure and volume of the heart chambers are measured by inserting a sensor inside the cavity and the impact of ischemia and/or drug effects can be evaluated. Detailed electrophysiological properties can be studied by applying electrodes and also by imaging the fluorescent probes that are sensitive to ion fluxes(116). The advantage of this method is that it avoids interference from the autonomic nerve system and inter-organ communications, which allows examination of pure cardiac response against various interventions. Cross-circulation method is another ex-vivo model system(117). By circulating the heart through the blood circulation from another animal, it avoids potential interference from the artificial perfusate, while more closely mimicking the pulsatile perfusion in in vivo setting.
4. In Vivo models
In contrast to the controlled and cardiac specific in vitro and ex vivo model systems, in vivo models offer studies in more complex biological system. In vivo heart function is regulated by multiple inputs including autonomic nervous system, secreted hormones, and immune systems. Thus, to understand the effects of pathological stimuli, genes, and drugs on the heart in a whole biological system, in vivo models are essential. Cardiac diseases have been modeled in drosophila(118), zebra fish(119), and xenopus(120) with a focus on gene regulation and development, but these models are beyond the scope of this review. We will review commonly used small and large animal models of cardiac disease in below.
4-1. Rodent models of cardiac diseases
Rodent models play central roles in the experimental cardiac research in the laboratories. They have 4-chamber cardiac anatomy similar to humans with high similarity in genomic sequences(121), and are relatively easy to handle, require smaller space and less costly compared to more advanced species. Mouse models have become the most popular owing to widely available genetically modified lines and established techniques to manipulate expression of genes. Rats are also commonly used in the laboratories and offer better surgical manipulations and imaging capabilities compared to mice. Genetically modified rat models are also increasingly used(122). Three major approaches to induce cardiac disease in rodents are surgical, pharmacological, and gene manipulation. Here, we summarize these models in below sections.
4-1a. Surgically induced models
Myocardial ischemia induced by surgical ligation or ischemic reperfusion of the coronary artery represents one of the most common surgical methods to model cardiac disease in rodents. Left anterior descending artery is usually targeted and a very thin suture is placed around the coronary artery. The coronary artery is ligated for inducing transmural infarction, whereas it is temporarily ligated and later released for ischemic reperfusion injury(123). Generally, 30 minutes of ischemia is sufficient to induce infarction, but the longer duration of ischemia and longer reperfusion time is associated with larger acute myocardial infarction(124). Successful induction of myocardial ischemia will result in tissue necrosis and apoptosis acutely, and leads to scar formation at the chronic stage. Degree of systolic dysfunction depends on size of the myocardial area perfused by the occluded coronary artery and ligation time until reperfusion(125). Challenges remain in inducing reproducible infarct size and cardiac dysfunction due to the inter-animal differences in coronary anatomy and difficulties in visualizing coronary arteries in some of the animals(126). Another commonly used surgically induced model is the aortic constriction. Aortic constriction increases cardiac afterload and the heart initially develops hypertrophy. Systolic function is usually maintained at this stage(127). However, after this adaptive hypertrophy phase, the heart gradually dilates and systolic function decreases(128). Degree of stenosis and the location of the constriction are the key factors that determine the speed of this process. Ascending aortic constriction generally results in more severe and rapid progression of heart failure, whereas it is slower with transverse aortic constriction. Commonly, 26 to 27 gauge needles are used to set the degree of stenosis for mouse transaortic constriction, while more variety of needle sizes are used for rat aortic constrictions. Type of sutures used can also affect the model phenotype as the sutures that swell (e.g. silk sutures) can expand and tighten the stenosis after surgery. Systolic dysfunction may not always develop in rat aortic constriction model and only part of the animals with severe ascending aortic constriction show decreased ejection fraction(129). Even using same tools and techniques, the speed of disease progression is sometimes operator dependent. Thus, prior characterization of operator-dependent disease progression is one of the key elements for designing the experimental study using this model. For inducing volume-overload heart failure, needle disruption of aortic(130) or mitral valve(131), and creation of aorto-venous fistula(132) are employed and result in dilation of the cardiac chambers with different cardiac remodeling pattern compared to pressure overload models as described later in the large animal model section(133).
4-1b. Pharmacologically induced models
Pharmacological approaches are also common in inducing cardiac diseases in rodents. Cardio toxins such as doxorubicin and trastuzumab are known to induce cardiac dysfunction in dose dependent manner. In addition to decreased cardiac contraction, interstitial fibrosis and increased cell death are found(134). Myocarditis are induced by injection of Coxsackievirus(135)or auto-antibodies against cardiomyocyte structures such as myosine(136) and troponin I(137). Myocarditis leads to dilated cardiomyopathy at chronic stages in these models(135). Continuous infusion of isoproterenol using implantable osmotic pump can induce hypertrophy and eventually systolic dysfunction(138,139). Angiotensin II infusion using same technique can result in hypertrophy with characteristics similar to heart failure with preserved ejection fraction(140,141), while combination of unilateral nephrectomy and salty diet led to impaired systolic function(142). Monocrotaline is another substance that is used very commonly in rats to induce pulmonary hypertension. Single injection of monocrotaline results in pulmonary vascular remodeling and right ventricular failure that has similar characteristics to clinical pulmonary arterial hypertension. Nevertheless, some of the drugs that have shown efficacy in this model failed to show same benefit in clinical trials, suggesting incomplete representation of human disease. In addition, despite its advantage on simple and reliable induction method, monocrotaline is not very effective in inducing severe pulmonary hypertension in mice. To induce pulmonary hypertension in mice, SU5416 (vascular endothelial growth factor receptor inhibitor) is often used in combination with hypoxia or other pulmonary vascular injury approaches. Although this is more cumbersome than monocrotaline model, it exhibits closer histological features to human pulmonary arterial hypertension(143).
4-1c. Gene manipulation models
Different cardiac phenotypes induced by specific gene knockout or overexpression are not within the scope of this review. There are some commonly used gene manipulated rodent models including salt-sensitive and spontaneously hypertensive mouse(144) and rat(145), obesity and type 2 diabetes(146), and combination of both hypertension and obesity(147). These models exhibit diastolic dysfunction and are important tools to study heart failure with preserved ejection fraction.
4-2. Other small and middle sized animal models
Guinea pigs, cats, and rabbits are also used for cardiac research. Their classifications are somewhat vague and guinea pigs are often included in rodents, and cats and rabbits are sometimes referred to as large animals. They are larger in size compared to mice and rats and offer easier surgical manipulation. Cardiac physiology is more similar to humans, as the heart rate is generally lower in larger species. In addition, guinea pigs present more similarity to human in electrophysiological properties compared to mice and rats(148), including intracellular Ca2+ handling, action potential shape, human-like QT alterations and arrhythmias associated with heart failure(149). Aortic banding is commonly performed in guinea pigs to induce hypertrophy(150). Cats with spontaneous hypertrophic cardiomyopathy have been reported(151) and provide opportunities to study this clinically relevant disease(152). Rabbits show similarities to human in myosin heavy chain ratio (β type predominant)(153) and abundance of calcium cycling proteins(154). Specifically, contribution of Na+-Ca2+ exchanger in diastolic Ca2+clearance is around 28–29% in rabbits, which is around 4-fold higher than mice and rats, while similar to humans(155). Surgical approaches are commonly used to induce cardiac diseases such as aortic banding, coronary ligation and tachypacing after pacemaker implantation(156). A unique rabbit model of vulnerable atherosclerotic plaque rapture was developed by a combination of vascular injury and high cholesterol diet(157). These middlesized animal models offer experiments in more physiologically closer conditions to humans while being relatively cost-efficient compared to the larger animal models described in the following section.
4-3. Large animal models of cardiac diseases
Large animals commonly used for cardiac research include pigs, dogs, sheep and non-human primates. Their advantages over smaller animal models are physiological relevance and size similarity to humans, closer molecular characteristics, and availability in testing clinical sized devices and catheters. The disadvantage includes high cost for acquiring and maintaining the animals, requirement of large space and man power, more ethical concerns, and difficulties in obtaining aged or transgenic animals. Nevertheless, large animal experiments are the key step for translating new therapeutic approaches toward clinic by testing the efficacy in clinically relevant species, examining the safety, and defining optimal endpoint for future clinical trials. Several models have been established each mimicking different cardiac disease phenotypes that are found in patients.
4-3a. Ischemia models
Ischemia induced cardiac disease models are most commonly used in large animal cardiac research for its reproducible induction of systolic dysfunction and relatively simple method. Myocardial infarction associated with ischemic reperfusion injury can be developed by temporally blocking the antegrade coronary flow followed by reperfusion. Duration of ischemia determines the transmurality of the infarct, and short ischemia only induces myocardial stunning(158). Hedström et al(159) reported that duration of ischemia to develop 50% of infarction in the area at risk is significantly different between the species. Dogs required significantly longer ischemic time (181 min) compared to pigs (37 min) and rats (41 min) suggesting the importance of coronary collaterals, which is inherently rich in dogs(160). Left anterior descending artery or the left circumflex artery is commonly targeted to induce left heart failure. Occlusion of the proximal part of the left anterior descending artery is associated with larger infarct size and more cardiac remodeling at the chronic stage compared to the proximal left circumflex occlusion in pigs(161). However, some species have large left circumflex artery and the size of infarct follows the size of the area perfused by the occluded coronary artery. Thus, angiogram prior to induction of myocardial infarction can help predict the size of infarction. In contrast to semi-transmural infarction, a complete transmural myocardial infarction can be induced by surgically ligating the coronary artery or occluding it with embolic coils using endovascular technique. Both approaches result in similar infarct size, but the degree of inflammation and vascularization may be more close to clinical MI after endovascular approach(162). Microembolization is another method that has been used to induce ischemic heart failure(163), but this method usually requires multiple injections of microbeads to induce modest cardiac dysfunction. Although cumbersome, advantages of this model are that the degree of dysfunction can be adjusted, and global LV dysfunction can be induced in contrast to the regional dysfunction seen in other MI models. Overdose injection of microbeads can be used to induce an acute cardiogenic shock model.
In patients, ischemic disease does not always develop acutely and slow progression of coronary atherosclerosis can cause chronic total occlusion. The ischemic myocardium is often rescued by collateral flow through small bridging channels, but the myocardium becomes dysfunctional due to insufficient blood supply. This dysfunctional ischemic myocardium remain viable and is commonly called hibernating myocardium. Models of hibernating myocardium have been developed to study this interesting myocardial phenotype using coronary occluders(164–166) and flow limiting stents(167).
4-3b. Pressure-overload model
In contrast to the frequent use of pressure-overload models in rodent studies, large animal models of pressure-overload are less commonly employed relative to ischemic models. Nevertheless, considering the wide prevalence of hypertension in the clinic, large animal models of pressure-overload is crucial. Similar to rodents, aortic banding can be applied to large animals using surgical techniques. For the ascending aortic banding model, the hemodynamic profile is similar to patients with aortic stenosis except for that coronary arteries are perfused with increased pressure in animal models of ascending aortic banding. Whether this difference influences heart failure phenotype has not been studied well. These models exhibit significant macroscopic and microscopic hypertrophy, but systolic dysfunction seems to progress much slower than rodent models, if it does(168,169). Heart failure with preserved ejection fraction is a common heart failure phenotype found in the clinic and patients often have hypertension, suggesting its association to cardiac pressure-overload. Development of animal models that completely recapitulate heart failure with preserved ejection fraction is challenging, as the disease pathophysiology is likely multifactorial(170,171). Kidney wrapping on aged dogs has been reported to exhibit similar cardiac phenotype to heart failure with preserved ejection fraction and probably is one of the most relevant large animal models of this disease(172). Specifically, the animals exhibit impaired relaxation, increased left ventricular stiffness, and left atrial remodeling together with histological hypertrophy and fibrosis of myocardium(172,173). There are also efforts to create new clinically relevant models by inducing metabolic diseases in large animals(174).
4-3c. Volume-overload model
Valvular regurgitation, left-to-right cardiac or vascular shunt, and anemia can overload the heart with excessive volume and can promote cardiac dysfunction. Increased volume load results in high left ventricular end-diastolic pressure and dilates all cardiac chambers. Generally, systolic function is maintained until the end-stage of heart failure, but the heart dilates and exhibits eccentric hypertrophy(175). Both aortic and mitral valves can be targeted to overload the LV, but mitral valve regurgitation has been more often induced in large animal models. Surgical or percutanous severing of mitral valve cordae tendinae reliably induces mitral regurgitation and leads to development of chronic heart failure, but is accompanied by high mortality as high as 50%(176,177). High mortality is likely related to the diffculty in controlling the degree of regurgitation in this method. A unique method to control the degree of left ventricle to left atrial regurgitation is to implant a graft between the left ventricule and the left atrium(178). Although implantation is technically challenging, clmping of the graft enables easy correction of the regurgitation. For acute induction of mitral regurgitation, placement of inferior vena cava filters has been used and the degree of regurgitation can be controlled by adjusing the expansion of the filter(179). Mitral regurgitation also developes in patients post-myocardial infarction and is a predictor of worse prognosis. This can be reproduced in large animals by creating large infarction in the left ventricular posterior wall in sheep and pigs. These ischemic mitral regurgitation models generally exhibit progressive deterioration of heart failure(180,181). Artery to venous fistula models are another common method to induce volume-overload heart failure in large animals(182). The degree of the shunt and the proximisity to the heart are the key factors that determine the severity of heart failure.
4-3d. Other phenotypes of heart failure
Tachycardia induced heart failure is a well-established and reproducible large animal model of non-ischemic cardiac failure(183,184). Rapid pacing of the atrium or the ventricle leads to elevation of left ventricular end-diastolic pressure accompanied by systolic dysfunction. Among different species, dog model of pacing induced tachycardiac heart failure has been the best characterized, and it uses a pacing rate of 200–230 bpm for 3–4 weeks. Slower pacing and shorter duration of pacing result in less severe heart failure phenotype(185). In this model, activation of neurohormonal systems and molecular changes relevant to human heart failure has been demonstrated(186). Similar to human tachycardic heart failure, cessation of tachypacing results in recovery of cardiac function, thus the dysfunction is reversible to a certain extent. Cardio-toxic drug injections into the coronary artery result in systolic dysfunction with fibrosis and myofiber atrophy(187,188). However, this approach is associated with high mortality and requires repetitive administration of drugs. In addition to the left heart failure, right heart failure can be induced by pulmonary arterial banding(189), pulmonary vein banding(190,191), pulmonary microbeads injection(192), artery-to-venous shunt(193) and pneumonectomy(194). In contrast to the LV failure models, right ventricular failure models are much less characterized and more research is needed to identify the differences between the right and left heart failures.
5. Conclusion
We reviewed commonly used in vitro and in vivo models of cardiac diseases. Wide spectrum of models offers various experiments dedicated to examine specific pathways and therapies, but the researchers need to be aware of their characteristics and relevance to clinical cardiac diseases. Development of more easily-induced, reproducible, and clinically relevant models as well as characterization of new and existing models are important for further refinement of experimental cardiac research.
Acknowledgments
This work is supported by AHA-SDG 17SDG33410873 (K.I.), and AHA 17POST33410877 (J.G.O.), NIH R01 HL139963 (K.I.), R00 HL116645 (C.K.), R01 HL119046, R01 HL117505, R01 HL128099, R01 HL129814, R01HL131404, & T32 HL007824 (R. J. H.), and a Transatlantic Leducq Foundation grant.
Footnotes
Conflict of Interest: Author J.O., Author C.K., Author R.J.H,and Author K.I. declare that they have no conflict of interest.
References
- 1.Ziaeian B, Fonarow GC. Epidemiology and aetiology of heart failure. Nat Rev Cardiol 2016;13:368–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Banerjee I, Fuseler JW, Price RL, Borg TK, Baudino TA. Determination of cell types and numbers during cardiac development in the neonatal and adult rat and mouse. Am J Physiol Heart Circ Physiol 2007;293:H1883–91. [DOI] [PubMed] [Google Scholar]
- 3.Fujiu K, Nagai R. Contributions of cardiomyocyte-cardiac fibroblast-immune cell interactions in heart failure development. Basic Res Cardiol 2013;108:357. [DOI] [PubMed] [Google Scholar]
- 4.Sequeira V, van der Velden J. Historical perspective on heart function: the Frank-Starling Law. Biophys Rev 2015;7:421–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Brette F, Orchard C. T-tubule function in mammalian cardiac myocytes. Circ Res 2003;92:1182–92. [DOI] [PubMed] [Google Scholar]
- 6.Chlopcikova S, Psotova J, Miketova P. Neonatal rat cardiomyocytes--a model for the study of morphological, biochemical and electrophysiological characteristics of the heart. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2001;145:49–55. [PubMed] [Google Scholar]
- 7.Muller-Werdan U, Klein D, Zander M, Werdan K, Hammer C. Beating neonatal rat cardiomyocytes as a model to study the role of xenoreactive natural antibodies in xenotransplantation. Transplantation 1994;58:1403–9. [PubMed] [Google Scholar]
- 8.Louch WE, Sheehan KA, Wolska BM. Methods in cardiomyocyte isolation, culture, and gene transfer. J Mol Cell Cardiol 2011;51:288–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Djurovic S, Iversen N, Jeansson S, Hoover F, Christensen G. Comparison of nonviral transfection and adeno-associated viral transduction on cardiomyocytes. Mol Biotechnol 2004;28:21–32. [DOI] [PubMed] [Google Scholar]
- 10.Frank D, Kuhn C, Brors B et al. Gene expression pattern in biomechanically stretched cardiomyocytes: evidence for a stretch-specific gene program. Hypertension 2008;51:309–18. [DOI] [PubMed] [Google Scholar]
- 11.Harary I, Farley B. In vitro studies on single beating rat heart cells. I. Growth and organization. Exp Cell Res 1963;29:451–65. [DOI] [PubMed] [Google Scholar]
- 12.Mohamed BA, Barakat AZ, Zimmermann WH et al. Targeted disruption of Hspa4 gene leads to cardiac hypertrophy and fibrosis. J Mol Cell Cardiol 2012;53:459–68. [DOI] [PubMed] [Google Scholar]
- 13.Simpson P, McGrath A, Savion S. Myocyte hypertrophy in neonatal rat heart cultures and its regulation by serum and by catecholamines. Circ Res 1982;51:787–801. [DOI] [PubMed] [Google Scholar]
- 14.Huang Q, Huang J, Zeng Z et al. Effects of ERK1/2/PPARalpha/SCAD signal pathways on cardiomyocyte hypertrophy induced by insulin-like growth factor 1 and phenylephrine. Life Sci 2015;124:41–9. [DOI] [PubMed] [Google Scholar]
- 15.Nakaoka M, Iwai-Kanai E, Katamura M, Okawa Y, Mita Y, Matoba S. An alpha-adrenergic agonist protects hearts by inducing Akt1-mediated autophagy. Biochem Biophys Res Commun 2015;456:250–6. [DOI] [PubMed] [Google Scholar]
- 16.Zobel C, Kassiri Z, Nguyen TT, Meng Y, Backx PH. Prevention of hypertrophy by overexpression of Kv4.2 in cultured neonatal cardiomyocytes. Circulation 2002;106:2385–91. [DOI] [PubMed] [Google Scholar]
- 17.Menaouar A, Florian M, Wang D, Danalache B, Jankowski M, Gutkowska J. Anti-hypertrophic effects of oxytocin in rat ventricular myocytes. Int J Cardiol 2014;175:38–49. [DOI] [PubMed] [Google Scholar]
- 18.Sadoshima J, Izumo S. Molecular characterization of angiotensin II--induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts. Critical role of the AT1 receptor subtype. Circ Res 1993;73:413–23. [DOI] [PubMed] [Google Scholar]
- 19.Sakai S, Shimojo N, Kimura T et al. Involvement of peptidyl-prolyl isomerase Pin1 in the inhibitory effect of fluvastatin on endothelin-1-induced cardiomyocyte hypertrophy. Life Sci 2014;102:98–104. [DOI] [PubMed] [Google Scholar]
- 20.Reid BG, Stratton MS, Bowers S et al. Discovery of novel small molecule inhibitors of cardiac hypertrophy using high throughput, high content imaging. J Mol Cell Cardiol 2016;97:106–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Skwarek-Maruszewska A, Hotulainen P, Mattila PK, Lappalainen P. Contractility-dependent actin dynamics in cardiomyocyte sarcomeres. J Cell Sci 2009;122:2119–26. [DOI] [PubMed] [Google Scholar]
- 22.Fan X, Hughes BG, Ali MA, Chan BY, Launier K, Schulz R. Matrix metalloproteinase-2 in oncostatin M-induced sarcomere degeneration in cardiomyocytes. Am J Physiol Heart Circ Physiol 2016;311:H183–9. [DOI] [PubMed] [Google Scholar]
- 23.Komuro I, Kaida T, Shibazaki Y et al. Stretching cardiac myocytes stimulates protooncogene expression. J Biol Chem 1990;265:3595–8. [PubMed] [Google Scholar]
- 24.Komuro I, Katoh Y, Kaida T et al. Mechanical loading stimulates cell hypertrophy and specific gene expression in cultured rat cardiac myocytes. Possible role of protein kinase C activation. J Biol Chem 1991;266:1265–8. [PubMed] [Google Scholar]
- 25.Lin L, Tang C, Xu J et al. Mechanical stress triggers cardiomyocyte autophagy through angiotensin II type 1 receptor-mediated p38MAP kinase independently of angiotensin II. PLoS One 2014;9:e89629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Choudhary R, Baker KM, Pan J. All-trans retinoic acid prevents angiotensin II- and mechanical stretch-induced reactive oxygen species generation and cardiomyocyte apoptosis. J Cell Physiol 2008;215:172–81. [DOI] [PubMed] [Google Scholar]
- 27.Cheng TH, Chen JJ, Shih NL et al. Mechanical stretch induces endothelial nitric oxide synthase gene expression in neonatal rat cardiomyocytes. Clin Exp Pharmacol Physiol 2009;36:559–66. [DOI] [PubMed] [Google Scholar]
- 28.Wang BW, Wu GJ, Cheng WP, Shyu KG. Mechanical stretch via transforming growth factor-beta1 activates microRNA-208a to regulate hypertrophy in cultured rat cardiac myocytes. J Formos Med Assoc 2013;112:635–43. [DOI] [PubMed] [Google Scholar]
- 29.Liu W, Wang X, Mei Z et al. Chronic stress promotes the progression of pressure overload-induced cardiac dysfunction through inducing more apoptosis and fibrosis. Physiol Res 2015;64:325–34. [DOI] [PubMed] [Google Scholar]
- 30.Acosta D, Puckett M. Ischemic myocardial injury in cultured heart cells: preliminary observations on morphology and beating activity. In Vitro 1977;13:818–23. [DOI] [PubMed] [Google Scholar]
- 31.Wang XX, Wang XL, Tong MM et al. SIRT6 protects cardiomyocytes against ischemia/reperfusion injury by augmenting FoxO3alpha-dependent antioxidant defense mechanisms. Basic Res Cardiol 2016;111:13. [DOI] [PubMed] [Google Scholar]
- 32.Peng K, Qiu Y, Li J, Zhang ZC, Ji FH. Dexmedetomidine attenuates hypoxia/reoxygenation injury in primary neonatal rat cardiomyocytes. Exp Ther Med 2017;14:689–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bagheri F, Khori V, Alizadeh AM, Khalighfard S, Khodayari S, Khodayari H. Reactive oxygen species-mediated cardiac-reperfusion injury: Mechanisms and therapies. Life Sci 2016;165:43–55. [DOI] [PubMed] [Google Scholar]
- 34.Diaz RJ, Wilson GJ. Studying ischemic preconditioning in isolated cardiomyocyte models. Cardiovasc Res 2006;70:286–96. [DOI] [PubMed] [Google Scholar]
- 35.Graham RM, Frazier DP, Thompson JW et al. A unique pathway of cardiac myocyte death caused by hypoxia-acidosis. J Exp Biol 2004;207:3189–200. [DOI] [PubMed] [Google Scholar]
- 36.Musters RJ, Post JA, Verkleij AJ. The isolated neonatal rat-cardiomyocyte used in an in vitro model for ‘ischemia’. I. A morphological study. Biochim Biophys Acta 1991;1091:270–7. [DOI] [PubMed] [Google Scholar]
- 37.Kim MY, Seo EJ, Lee DH et al. Gadd45beta is a novel mediator of cardiomyocyte apoptosis induced by ischaemia/hypoxia. Cardiovasc Res 2010;87:119–26. [DOI] [PubMed] [Google Scholar]
- 38.Tu S, Liu ZQ, Fu JJ, Zhu WF, Luo DY, Wan FS. Inhibitory effect of p53 upregulated modulator of apoptosis targeting siRNA on hypoxia/reoxygenation-induced cardiomyocyte apoptosis in rats. Cardiology 2012;122:93–100. [DOI] [PubMed] [Google Scholar]
- 39.Gilsbach R, Preissl S, Gruning BA et al. Dynamic DNA methylation orchestrates cardiomyocyte development, maturation and disease. Nat Commun 2014;5:5288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dolinsky VW, Soltys CL, Rogan KJ et al. Resveratrol prevents pathological but not physiological cardiac hypertrophy. J Mol Med (Berl) 2015;93:413–25. [DOI] [PubMed] [Google Scholar]
- 41.von Lueder TG, Wang BH, Kompa AR et al. Angiotensin receptor neprilysin inhibitor LCZ696 attenuates cardiac remodeling and dysfunction after myocardial infarction by reducing cardiac fibrosis and hypertrophy. Circ Heart Fail 2015;8:71–8. [DOI] [PubMed] [Google Scholar]
- 42.Bell RM, Mocanu MM, Yellon DM. Retrograde heart perfusion: the Langendorff technique of isolated heart perfusion. J Mol Cell Cardiol 2011;50:940–50. [DOI] [PubMed] [Google Scholar]
- 43.Tan T, Marin-Garcia J, Damle S, Weiss HR. Hypoxia-inducible factor-1 improves inotropic responses of cardiac myocytes in ageing heart without affecting mitochondrial activity. Exp Physiol 2010;95:712–22. [DOI] [PubMed] [Google Scholar]
- 44.Mellor KM, Curl CL, Chandramouli C, Pedrazzini T, Wendt IR, Delbridge LM. Ageing-related cardiomyocyte functional decline is sex and angiotensin II dependent. Age (Dordr) 2014;36:9630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang YX, Korth M. Effects of doxorubicin on excitation-contraction coupling in guinea pig ventricular myocardium. Circ Res 1995;76:645–53. [DOI] [PubMed] [Google Scholar]
- 46.Oh JG, Kim J, Jang SP et al. Decoy peptides targeted to protein phosphatase 1 inhibit dephosphorylation of phospholamban in cardiomyocytes. J Mol Cell Cardiol 2013;56:63–71. [DOI] [PubMed] [Google Scholar]
- 47.Zhang M, Prosser BL, Bamboye MA et al. Contractile Function During Angiotensin-II Activation: Increased Nox2 Activity Modulates Cardiac Calcium Handling via Phospholamban Phosphorylation. J Am Coll Cardiol 2015;66:261–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Toischer K, Zhu W, Hunlich M et al. Cardiomyocyte proliferation prevents failure in pressure overload but not volume overload. J Clin Invest 2017;127:4285–4296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Du CK, Morimoto S, Nishii K et al. Knock-in mouse model of dilated cardiomyopathy caused by troponin mutation. Circ Res 2007;101:185–94. [DOI] [PubMed] [Google Scholar]
- 50.Inoue T, Kobirumaki-Shimozawa F, Kagemoto T et al. Depressed Frank-Starling mechanism in the left ventricular muscle of the knock-in mouse model of dilated cardiomyopathy with troponin T deletion mutation DeltaK210. J Mol Cell Cardiol 2013;63:69–78. [DOI] [PubMed] [Google Scholar]
- 51.Hu LR, Ackermann MA, Hecker PA et al. Deregulated Ca(2+) cycling underlies the development of arrhythmia and heart disease due to mutant obscurin. Sci Adv 2017;3:e1603081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bhargava A, Lin X, Novak P et al. Super-resolution scanning patch clamp reveals clustering of functional ion channels in adult ventricular myocyte. Circ Res 2013;112:1112–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gaitas A, Malhotra R, Li T, Herron T, Jalife J. A device for rapid and quantitative measurement of cardiac myocyte contractility. Rev Sci Instrum 2015;86:034302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Moshal KS, Tipparaju SM, Vacek TP et al. Mitochondrial matrix metalloproteinase activation decreases myocyte contractility in hyperhomocysteinemia. Am J Physiol Heart Circ Physiol 2008;295:H890–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cagalinec M, Waczulikova I, Ulicna O, Chorvat D Jr. Morphology and contractility of cardiac myocytes in early stages of streptozotocin-induced diabetes mellitus in rats. Physiol Res 2013;62:489–501. [DOI] [PubMed] [Google Scholar]
- 56.Kerr JP, Robison P, Shi G et al. Detyrosinated microtubules modulate mechanotransduction in heart and skeletal muscle. Nat Commun 2015;6:8526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Thandapilly SJ, Louis XL, Yang T et al. Resveratrol prevents norepinephrine induced hypertrophy in adult rat cardiomyocytes, by activating NO-AMPK pathway. Eur J Pharmacol 2011;668:217–24. [DOI] [PubMed] [Google Scholar]
- 58.Eom GH, Nam YS, Oh JG et al. Regulation of acetylation of histone deacetylase 2 by p300/CBP-associated factor/histone deacetylase 5 in the development of cardiac hypertrophy. Circ Res 2014;114:1133–43. [DOI] [PubMed] [Google Scholar]
- 59.Sowah D, Brown BF, Quon A, Alvarez BV, Casey JR. Resistance to cardiomyocyte hypertrophy in ae3−/− mice, deficient in the AE3 Cl-/HCO3- exchanger. BMC Cardiovasc Disord 2014;14:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Miller CL, Oikawa M, Cai Y et al. Role of Ca2+/calmodulin-stimulated cyclic nucleotide phosphodiesterase 1 in mediating cardiomyocyte hypertrophy. Circ Res 2009;105:956–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nam YS, Kim Y, Joung H et al. Small heterodimer partner blocks cardiac hypertrophy by interfering with GATA6 signaling. Circ Res 2014;115:493–503. [DOI] [PubMed] [Google Scholar]
- 62.Iribe G, Kohl P. Axial stretch enhances sarcoplasmic reticulum Ca2+ leak and cellular Ca2+ reuptake in guinea pig ventricular myocytes: experiments and models. Prog Biophys Mol Biol 2008;97:298–311. [DOI] [PubMed] [Google Scholar]
- 63.Prosser BL, Ward CW, Lederer WJ. X-ROS signalling is enhanced and graded by cyclic cardiomyocyte stretch. Cardiovasc Res 2013;98:307–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Marvin WJ Jr., Robinson RB, Hermsmeyer K. Correlation of function and morphology of neonatal rat and embryonic chick cultured cardiac and vascular muscle cells. Circ Res 1979;45:528–40. [DOI] [PubMed] [Google Scholar]
- 65.Claycomb WC, Palazzo MC. Culture of the terminally differentiated adult cardiac muscle cell: a light and scanning electron microscope study. Dev Biol 1980;80:466–82. [DOI] [PubMed] [Google Scholar]
- 66.Kimes BW, Brandt BL. Properties of a clonal muscle cell line from rat heart. Exp Cell Res 1976;98:367–81. [DOI] [PubMed] [Google Scholar]
- 67.Steinhelper ME, Lanson NA Jr., Dresdner KP et al. Proliferation in vivo and in culture of differentiated adult atrial cardiomyocytes from transgenic mice. Am J Physiol 1990;259:H1826–34. [DOI] [PubMed] [Google Scholar]
- 68.Delcarpio JB, Lanson NA Jr., Field LJ, Claycomb WC. Morphological characterization of cardiomyocytes isolated from a transplantable cardiac tumor derived from transgenic mouse atria (AT-1 cells). Circ Res 1991;69:1591–600. [DOI] [PubMed] [Google Scholar]
- 69.Jaffredo T, Chestier A, Bachnou N, Dieterlen-Lievre F. MC29-immortalized clonal avian heart cell lines can partially differentiate in vitro. Exp Cell Res 1991;192:481–91. [DOI] [PubMed] [Google Scholar]
- 70.Claycomb WC, Lanson NA Jr., Stallworth BS et al. HL-1 cells: a cardiac muscle cell line that contracts and retains phenotypic characteristics of the adult cardiomyocyte. Proc Natl Acad Sci U S A 1998;95:2979–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Davidson MM, Nesti C, Palenzuela L et al. Novel cell lines derived from adult human ventricular cardiomyocytes. J Mol Cell Cardiol 2005;39:133–47. [DOI] [PubMed] [Google Scholar]
- 72.Menard C, Pupier S, Mornet D, Kitzmann M, Nargeot J, Lory P. Modulation of L-type calcium channel expression during retinoic acid-induced differentiation of H9C2 cardiac cells. J Biol Chem 1999;274:29063–70. [DOI] [PubMed] [Google Scholar]
- 73.Branco AF, Pereira SP, Gonzalez S, Gusev O, Rizvanov AA, Oliveira PJ. Gene Expression Profiling of H9c2 Myoblast Differentiation towards a Cardiac-Like Phenotype. PLoS One 2015;10:e0129303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Watkins SJ, Borthwick GM, Arthur HM. The H9C2 cell line and primary neonatal cardiomyocyte cells show similar hypertrophic responses in vitro. In Vitro Cell Dev Biol Anim 2011;47:125–31. [DOI] [PubMed] [Google Scholar]
- 75.Wang G, Tang C, Yan G, Feng B. Gene Expression Profiling of H9c2 Cells Subjected to H2O2-Induced Apoptosis with/without AF-HF001. Biol Pharm Bull 2016;39:207–14. [DOI] [PubMed] [Google Scholar]
- 76.Kuznetsov AV, Javadov S, Sickinger S, Frotschnig S, Grimm M. H9c2 and HL-1 cells demonstrate distinct features of energy metabolism, mitochondrial function and sensitivity to hypoxiareoxygenation. Biochim Biophys Acta 2015;1853:276–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Branco AF, Pereira SL, Moreira AC, Holy J, Sardao VA, Oliveira PJ. Isoproterenol cytotoxicity is dependent on the differentiation state of the cardiomyoblast H9c2 cell line. Cardiovasc Toxicol 2011;11:191–203. [DOI] [PubMed] [Google Scholar]
- 78.McWhinney CD, Hansen C, Robishaw JD. Alpha-1 adrenergic signaling in a cardiac murine atrial myocyte (HL-1) cell line. Mol Cell Biochem 2000;214:111–9. [DOI] [PubMed] [Google Scholar]
- 79.Kitta K, Clement SA, Remeika J, Blumberg JB, Suzuki YJ. Endothelin-1 induces phosphorylation of GATA-4 transcription factor in the HL-1 atrial-muscle cell line. Biochem J 2001;359:375–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Haas S, Jahnke HG, Moerbt N et al. DIGE proteome analysis reveals suitability of ischemic cardiac in vitro model for studying cellular response to acute ischemia and regeneration. PLoS One 2012;7:e31669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hong JH, Choi JH, Kim TY, Lee KJ. Spiral reentry waves in confluent layer of HL-1 cardiomyocyte cell lines. Biochem Biophys Res Commun 2008;377:1269–73. [DOI] [PubMed] [Google Scholar]
- 82.Sartiani L, Bochet P, Cerbai E, Mugelli A, Fischmeister R. Functional expression of the hyperpolarization-activated, non-selective cation current I(f) in immortalized HL-1 cardiomyocytes. J Physiol 2002;545:81–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dias P, Desplantez T, El-Harasis MA et al. Characterisation of connexin expression and electrophysiological properties in stable clones of the HL-1 myocyte cell line. PLoS One 2014;9:e90266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Fang X, Robinson J, Wang-Hu J et al. cAMP induces hypertrophy and alters DNA methylation in HL-1 cardiomyocytes. Am J Physiol Cell Physiol 2015;309:C425–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bloch L, Ndongson-Dongmo B, Kusch A, Dragun D, Heller R, Huber O. Real-time monitoring of hypertrophy in HL-1 cardiomyocytes by impedance measurements reveals different modes of growth. Cytotechnology 2016;68:1897–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Asensio-Lopez MC, Soler F, Pascual-Figal D, Fernandez-Belda F, Lax A. Doxorubicin-induced oxidative stress: The protective effect of nicorandil on HL-1 cardiomyocytes. PLoS One 2017;12:e0172803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Dutta D, Xu J, Kim JS, Dunn WA Jr., Leeuwenburgh C. Upregulated autophagy protects cardiomyocytes from oxidative stress-induced toxicity. Autophagy 2013;9:328–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Truong J, Mailloux RJ, Chan HM. Impact of methylmercury exposure on mitochondrial energetics in AC16 and H9C2 cardiomyocytes. Toxicol In Vitro 2015;29:953–61. [DOI] [PubMed] [Google Scholar]
- 89.Li Q, Qi X, Jia W. 3,3’,5-triiodothyroxine inhibits apoptosis and oxidative stress by the PKM2/PKM1 ratio during oxygen-glucose deprivation/reperfusion AC16 and HCM-a cells: T3 inhibits apoptosis and oxidative stress by PKM2/PKM1 ratio. Biochem Biophys Res Commun 2016;475:51–6. [DOI] [PubMed] [Google Scholar]
- 90.Cui L, Guo J, Zhang Q et al. Erythropoietin activates SIRT1 to protect human cardiomyocytes against doxorubicin-induced mitochondrial dysfunction and toxicity. Toxicol Lett 2017;275:28–38. [DOI] [PubMed] [Google Scholar]
- 91.Xiao Y, Yang Z, Wu QQ et al. Cucurbitacin B Protects Against Pressure Overload Induced Cardiac Hypertrophy. J Cell Biochem 2017;118:3899–3910. [DOI] [PubMed] [Google Scholar]
- 92.Thomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145–7. [DOI] [PubMed] [Google Scholar]
- 93.Messina E, De Angelis L, Frati G et al. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ Res 2004;95:911–21. [DOI] [PubMed] [Google Scholar]
- 94.Goumans MJ, de Boer TP, Smits AM et al. TGF-beta1 induces efficient differentiation of human cardiomyocyte progenitor cells into functional cardiomyocytes in vitro. Stem Cell Res 2007;1:138–49. [DOI] [PubMed] [Google Scholar]
- 95.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–76. [DOI] [PubMed] [Google Scholar]
- 96.Peters AK, Wouwer GV, Weyn B, Verheyen GR, Vanparys P, Gompel JV. Automated analysis of contractility in the embryonic stem cell test, a novel approach to assess embryotoxicity. Toxicol In Vitro 2008;22:1948–56. [DOI] [PubMed] [Google Scholar]
- 97.Liu H, Ren C, Liu W et al. Embryotoxicity estimation of commonly used compounds with embryonic stem cell test. Mol Med Rep 2017;16:263–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Burridge PW, Li YF, Matsa E et al. Human induced pluripotent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat Med 2016;22:547–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Maillet A, Tan K, Chai X et al. Modeling Doxorubicin-Induced Cardiotoxicity in Human Pluripotent Stem Cell Derived-Cardiomyocytes. Sci Rep 2016;6:25333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Seewald MJ, Ellinghaus P, Kassner A et al. Genomic profiling of developing cardiomyocytes from recombinant murine embryonic stem cells reveals regulation of transcription factor clusters. Physiol Genomics 2009;38:7–15. [DOI] [PubMed] [Google Scholar]
- 101.Shinozawa T, Tsuji A, Imahashi K et al. Gene expression profiling of functional murine embryonic stem cell-derived cardiomyocytes and comparison with adult heart: profiling of murine ESC-derived cardiomyocytes. J Biomol Screen 2009;14:239–45. [DOI] [PubMed] [Google Scholar]
- 102.Lee YK, Ng KM, Lai WH et al. Calcium homeostasis in human induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Rev 2011;7:976–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Carvajal-Vergara X, Sevilla A, D’Souza SL et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 2010;465:808–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wu H, Lee J, Vincent LG et al. Epigenetic Regulation of Phosphodiesterases 2A and 3A Underlies Compromised beta-Adrenergic Signaling in an iPSC Model of Dilated Cardiomyopathy. Cell Stem Cell 2015;17:89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lin B, Li Y, Han L et al. Modeling and study of the mechanism of dilated cardiomyopathy using induced pluripotent stem cells derived from individuals with Duchenne muscular dystrophy. Dis Model Mech 2015;8:457–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Crombie DE, Curl CL, Raaijmakers AJ et al. Friedreich’s ataxia induced pluripotent stem cell-derived cardiomyocytes display electrophysiological abnormalities and calcium handling deficiency. Aging (Albany NY) 2017;9:1440–1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Itzhaki I, Maizels L, Huber I et al. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll Cardiol 2012;60:990–1000. [DOI] [PubMed] [Google Scholar]
- 108.Guhr A, Kobold S, Seltmann S, Seiler Wulczyn AEM, Kurtz A, Loser P. Recent Trends in Research with Human Pluripotent Stem Cells: Impact of Research and Use of Cell Lines in Experimental Research and Clinical Trials. Stem Cell Reports 2018;11:485–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Mayourian J, Cashman TJ, Ceholski DK et al. Experimental and Computational Insight Into Human Mesenchymal Stem Cell Paracrine Signaling and Heterocellular Coupling Effects on Cardiac Contractility and Arrhythmogenicity. Circ Res 2017;121:411–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Grosberg A, Alford PW, McCain ML, Parker KK. Ensembles of engineered cardiac tissues for physiological and pharmacological study: heart on a chip. Lab Chip 2011;11:4165–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lee J, Razu ME, Wang X, Lacerda C, Kim JJ. Biomimetic cardiac microsystems for pathophysiological studies and drug screens. J Lab Autom 2015;20:96–106. [DOI] [PubMed] [Google Scholar]
- 112.Tanaka Y, Sato K, Shimizu T, Yamato M, Okano T, Kitamori T. A micro-spherical heart pump powered by cultured cardiomyocytes. Lab Chip 2007;7:207–12. [DOI] [PubMed] [Google Scholar]
- 113.Li RA, Keung W, Cashman TJ et al. Bioengineering an electro-mechanically functional miniature ventricular heart chamber from human pluripotent stem cells. Biomaterials 2018;163:116–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Olejnickova V, Novakova M, Provaznik I. Isolated heart models: cardiovascular system studies and technological advances. Med Biol Eng Comput 2015;53:669–78. [DOI] [PubMed] [Google Scholar]
- 115.de Bakker JM, Coronel R, Tasseron S et al. Ventricular tachycardia in the infarcted, Langendorffperfused human heart: role of the arrangement of surviving cardiac fibers. J Am Coll Cardiol 1990;15:1594–607. [DOI] [PubMed] [Google Scholar]
- 116.Akar JG, Akar FG. Mapping arrhythmias in the failing heart: from Langendorff to patient. J Electrocardiol 2006;39:S19–23. [DOI] [PubMed] [Google Scholar]
- 117.Takewa Y, Chemaly ER, Takaki M et al. Mechanical work and energetic analysis of eccentric cardiac remodeling in a volume overload heart failure in rats. Am J Physiol Heart Circ Physiol 2009;296:H1117–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Piazza N, Wessells RJ. Drosophila models of cardiac disease. Prog Mol Biol Transl Sci 2011;100:155–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhu XY, Wu SQ, Guo SY et al. A Zebrafish Heart Failure Model for Assessing Therapeutic Agents. Zebrafish 2018;15:243–253. [DOI] [PubMed] [Google Scholar]
- 120.Hempel A, Kuhl M. A Matter of the Heart: The African Clawed Frog Xenopus as a Model for Studying Vertebrate Cardiogenesis and Congenital Heart Defects. J Cardiovasc Dev Dis 2016;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Mouse Genome Sequencing C, Waterston RH, Lindblad-Toh K et al. Initial sequencing and comparative analysis of the mouse genome. Nature 2002;420:520–62. [DOI] [PubMed] [Google Scholar]
- 122.Flister MJ, Prokop JW, Lazar J et al. 2015 Guidelines for Establishing Genetically Modified Rat Models for Cardiovascular Research. J Cardiovasc Transl Res 2015;8:269–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gehrmann J, Frantz S, Maguire CT et al. Electrophysiological characterization of murine myocardial ischemia and infarction. Basic Res Cardiol 2001;96:237–50. [DOI] [PubMed] [Google Scholar]
- 124.Redel A, Jazbutyte V, Smul TM et al. Impact of ischemia and reperfusion times on myocardial infarct size in mice in vivo. Exp Biol Med 2008;233:84–93. [DOI] [PubMed] [Google Scholar]
- 125.Christia P, Bujak M, Gonzalez-Quesada C et al. Systematic characterization of myocardial inflammation, repair, and remodeling in a mouse model of reperfused myocardial infarction. J Histochem Cytochem 2013;61:555–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Chen J, Ceholski DK, Liang L, Fish K, Hajjar RJ. Variability in coronary artery anatomy affects consistency of cardiac damage after myocardial infarction in mice. Am J Physiol Heart Circ Physiol 2017;313:H275–H282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Lee A, Jeong D, Mitsuyama S et al. The role of SUMO-1 in cardiac oxidative stress and hypertrophy. Antioxid Redox Signal 2014;21:1986–2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hampton C, Rosa R, Campbell B et al. Early echocardiographic predictors of outcomes in the mouse transverse aortic constriction heart failure model. J Pharmacol Toxicol Methods 2017;84:93–101. [DOI] [PubMed] [Google Scholar]
- 129.Chaanine AH, Jeong D, Liang L et al. JNK modulates FOXO3a for the expression of the mitochondrial death and mitophagy marker BNIP3 in pathological hypertrophy and in heart failure. Cell Death Dis 2012;3:265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.You J, Wu J, Zhang Q et al. Differential cardiac hypertrophy and signaling pathways in pressure versus volume overload. Am J Physiol Heart Circ Physiol 2018;314:H552–H562. [DOI] [PubMed] [Google Scholar]
- 131.Pu M, Gao Z, Zhang X et al. Impact of mitral regurgitation on left ventricular anatomic and molecular remodeling and systolic function: implication for outcome. Am J Physiol Heart Circ Physiol 2009;296:H1727–32. [DOI] [PubMed] [Google Scholar]
- 132.Melenovsky V, Skaroupkova P, Benes J, Torresova V, Kopkan L, Cervenka L. The course of heart failure development and mortality in rats with volume overload due to aorto-caval fistula. Kidney Blood Press Res 2012;35:167–73. [DOI] [PubMed] [Google Scholar]
- 133.Chemaly ER, Kang S, Zhang S et al. Differential patterns of replacement and reactive fibrosis in pressure and volume overload are related to the propensity for ischaemia and involve resistin. J Physiol 2013;591:5337–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Angsutararux P, Luanpitpong S, Issaragrisil S. Chemotherapy-Induced Cardiotoxicity: Overview of the Roles of Oxidative Stress. Oxid Med Cell Longev 2015;2015:795602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Muller AM, Fischer A, Katus HA, Kaya Z. Mouse models of autoimmune diseases - autoimmune myocarditis. Curr Pharm Des 2015;21:2498–512. [DOI] [PubMed] [Google Scholar]
- 136.Pummerer CL, Luze K, Grassl G et al. Identification of cardiac myosin peptides capable of inducing autoimmune myocarditis in BALB/c mice. J Clin Invest 1996;97:2057–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kaya Z, Goser S, Buss SJ et al. Identification of cardiac troponin I sequence motifs leading to heart failure by induction of myocardial inflammation and fibrosis. Circulation 2008;118:2063–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Takeshita D, Shimizu J, Kitagawa Y et al. Isoproterenol-induced hypertrophied rat hearts: does short-term treatment correspond to long-term treatment? J Physiol Sci 2008;58:179–88. [DOI] [PubMed] [Google Scholar]
- 139.Wang JJ, Rau C, Avetisyan R et al. Genetic Dissection of Cardiac Remodeling in an Isoproterenol-Induced Heart Failure Mouse Model. PLoS Genet 2016;12:e1006038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Shinohara K, Kishi T, Hirooka Y, Sunagawa K. Circulating angiotensin II deteriorates left ventricular function with sympathoexcitation via brain angiotensin II receptor. Physiol Rep 2015;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Regan JA, Mauro AG, Carbone S et al. A mouse model of heart failure with preserved ejection fraction due to chronic infusion of a low subpressor dose of angiotensin II. Am J Physiol Heart Circ Physiol 2015;309:H771–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Tsukamoto Y, Mano T, Sakata Y et al. A novel heart failure mice model of hypertensive heart disease by angiotensin II infusion, nephrectomy, and salt loading. Am J Physiol Heart Circ Physiol 2013;305:H1658–67. [DOI] [PubMed] [Google Scholar]
- 143.Abe K, Toba M, Alzoubi A et al. Formation of plexiform lesions in experimental severe pulmonary arterial hypertension. Circulation 2010;121:2747–54. [DOI] [PubMed] [Google Scholar]
- 144.Lovelock JD, Monasky MM, Jeong EM et al. Ranolazine improves cardiac diastolic dysfunction through modulation of myofilament calcium sensitivity. Circ Res 2012;110:841–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Doi R, Masuyama T, Yamamoto K et al. Development of different phenotypes of hypertensive heart failure: systolic versus diastolic failure in Dahl salt-sensitive rats. J Hypertens 2000;18:111–20. [DOI] [PubMed] [Google Scholar]
- 146.Van den Bergh A, Vanderper A, Vangheluwe P et al. Dyslipidaemia in type II diabetic mice does not aggravate contractile impairment but increases ventricular stiffness. Cardiovasc Res 2008;77:371–9. [DOI] [PubMed] [Google Scholar]
- 147.Molinar-Toribio E, Perez-Jimenez J, Ramos-Romero S et al. Cardiovascular disease-related parameters and oxidative stress in SHROB rats, a model for metabolic syndrome. PLoS One 2014;9:e104637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Rosati B, Dong M, Cheng L et al. Evolution of ventricular myocyte electrophysiology. Physiol Genomics 2008;35:262–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Foster DB, Liu T, Kammers K et al. Integrated Omic Analysis of a Guinea Pig Model of Heart Failure and Sudden Cardiac Death. J Proteome Res 2016;15:3009–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Laviolle B, Pape D, Verdier MC, Lavenu A, Bellissant E. Hemodynamic and histomorphometric characteristics of heart failure induced by aortic stenosis in the guinea pig: comparison of two constriction sizes. Can J Physiol Pharmacol 2009;87:908–14. [DOI] [PubMed] [Google Scholar]
- 151.Fox PR, Basso C, Thiene G, Maron BJ. Spontaneously occurring restrictive nonhypertrophied cardiomyopathy in domestic cats: a new animal model of human disease. Cardiovasc Pathol 2014;23:28–34. [DOI] [PubMed] [Google Scholar]
- 152.Freeman LM, Rush JE, Stern JA, Huggins GS, Maron MS. Feline Hypertrophic Cardiomyopathy: A Spontaneous Large Animal Model of Human HCM. Cardiol Res 2017;8:139–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Suzuki T, Palmer BM, James J et al. Effects of cardiac myosin isoform variation on myofilament function and crossbridge kinetics in transgenic rabbits. Circ Heart Fail 2009;2:334–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Piacentino V 3rd, Weber CR, Chen X et al. Cellular basis of abnormal calcium transients of failing human ventricular myocytes. Circ Res 2003;92:651–8. [DOI] [PubMed] [Google Scholar]
- 155.Edwards AG, Louch WE. Species-Dependent Mechanisms of Cardiac Arrhythmia: A Cellular Focus. Clin Med Insights Cardiol 2017;11:1179546816686061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Freeman GL, Colston JT. Myocardial depression produced by sustained tachycardia in rabbits. Am J Physiol 1992;262:H63–7. [DOI] [PubMed] [Google Scholar]
- 157.Shimizu T, Nakai K, Morimoto Y et al. Simple rabbit model of vulnerable atherosclerotic plaque. Neurol Med Chir (Tokyo) 2009;49:327–32; [DOI] [PubMed] [Google Scholar]
- 158.Thomas SA, Fallavollita JA, Suzuki G, Borgers M, Canty JM Jr. Dissociation of regional adaptations to ischemia and global myolysis in an accelerated Swine model of chronic hibernating myocardium. Circ Res 2002;91:970–7. [DOI] [PubMed] [Google Scholar]
- 159.Hedstrom E, Engblom H, Frogner F et al. Infarct evolution in man studied in patients with first-time coronary occlusion in comparison to different species - implications for assessment of myocardial salvage. J Cardiovasc Magn Reson 2009;11:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Maxwell MP, Hearse DJ, Yellon DM. Species variation in the coronary collateral circulation during regional myocardial ischaemia: a critical determinant of the rate of evolution and extent of myocardial infarction. Cardiovasc Res 1987;21:737–46. [DOI] [PubMed] [Google Scholar]
- 161.Ishikawa K, Aguero J, Tilemann L et al. Characterizing preclinical models of ischemic heart failure: differences between LAD and LCx infarctions. Am J Physiol Heart Circ Physiol 2014;307:H1478–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Galvez-Monton C, Prat-Vidal C, Diaz-Guemes I et al. Comparison of two preclinical myocardial infarct models: coronary coil deployment versus surgical ligation. J Transl Med 2014;12:137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Saavedra WF, Tunin RS, Paolocci N et al. Reverse remodeling and enhanced adrenergic reserve from passive external support in experimental dilated heart failure. J Am Coll Cardiol 2002;39:2069–76. [DOI] [PubMed] [Google Scholar]
- 164.Page BJ, Banas MD, Suzuki G et al. Revascularization of chronic hibernating myocardium stimulates myocyte proliferation and partially reverses chronic adaptations to ischemia. J Am Coll Cardiol 2015;65:684–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Ishikawa K, Ladage D, Takewa Y et al. Development of a preclinical model of ischemic cardiomyopathy in swine. Am J Physiol Heart Circ Physiol 2011;301:H530–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Tuzun E, Oliveira E, Narin C et al. Correlation of ischemic area and coronary flow with ameroid size in a porcine model. J Surg Res 2010;164:38–42. [DOI] [PubMed] [Google Scholar]
- 167.Hinkel R, Howe A, Renner S et al. Diabetes Mellitus-Induced Microvascular Destabilization in the Myocardium. J Am Coll Cardiol 2017;69:131–143. [DOI] [PubMed] [Google Scholar]
- 168.Wisenbaugh T, Allen P, Cooper Gt, Holzgrefe H, Beller G, Carabello B. Contractile function, myosin ATPase activity and isozymes in the hypertrophied pig left ventricle after a chronic progressive pressure overload. Circ Res 1983;53:332–41. [DOI] [PubMed] [Google Scholar]
- 169.Ishikawa K, Aguero J, Oh JG et al. Increased stiffness is the major early abnormality in a pig model of severe aortic stenosis and predisposes to congestive heart failure in the absence of systolic dysfunction. J Am Heart Assoc 2015;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA. Evidence Supporting the Existence of a Distinct Obese Phenotype of Heart Failure With Preserved Ejection Fraction. Circulation 2017;136:6–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Borlaug BA. The pathophysiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2014;11:507–15. [DOI] [PubMed] [Google Scholar]
- 172.Zakeri R, Moulay G, Chai Q et al. Left Atrial Remodeling and Atrioventricular Coupling in a Canine Model of Early Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2016;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Munagala VK, Hart CY, Burnett JC Jr., Meyer DM, Redfield MM. Ventricular structure and function in aged dogs with renal hypertension: a model of experimental diastolic heart failure. Circulation 2005;111:1128–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Schwarzl M, Hamdani N, Seiler S et al. A porcine model of hypertensive cardiomyopathy: implications for heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2015;309:H1407–18. [DOI] [PubMed] [Google Scholar]
- 175.Opie LH, Commerford PJ, Gersh BJ, Pfeffer MA. Controversies in ventricular remodelling. Lancet 2006;367:356–67. [DOI] [PubMed] [Google Scholar]
- 176.Leroux AA, Moonen ML, Pierard LA, Kolh P, Amory H. Animal models of mitral regurgitation induced by mitral valve chordae tendineae rupture. J Heart Valve Dis 2012;21:416–23. [PubMed] [Google Scholar]
- 177.Watanabe S, Ishikawa K, Fish K et al. Protein Phosphatase Inhibitor-1 Gene Therapy in a Swine Model of Nonischemic Heart Failure. J Am Coll Cardiol 2017;70:1744–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Beeri R, Yosefy C, Guerrero JL et al. Mitral regurgitation augments post-myocardial infarction remodeling failure of hypertrophic compensation. J Am Coll Cardiol 2008;51:476–86. [DOI] [PubMed] [Google Scholar]
- 179.Naito N, Nishimura T, Takewa Y et al. What Is the Optimal Setting for a Continuous-Flow Left Ventricular Assist Device in Severe Mitral Regurgitation? Artif Organs 2016;40:1039–1045. [DOI] [PubMed] [Google Scholar]
- 180.Chaput M, Handschumacher MD, Guerrero JL et al. Mitral leaflet adaptation to ventricular remodeling: prospective changes in a model of ischemic mitral regurgitation. Circulation 2009;120:S99–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Ishikawa K, Watanabe S, Hammoudi N et al. Reduced longitudinal contraction is associated with ischemic mitral regurgitation after posterior MI. Am J Physiol Heart Circ Physiol 2018;314:H322–H329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Lu X, Zhang ZD, Guo X et al. Response of various conduit arteries in tachycardia- and volume overload-induced heart failure. PLoS One 2014;9:e101645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Woitek F, Zentilin L, Hoffman NE et al. Intracoronary Cytoprotective Gene Therapy: A Study of VEGF-B167 in a Pre-Clinical Animal Model of Dilated Cardiomyopathy. J Am Coll Cardiol 2015;66:139–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Spinale FG, Coker ML, Thomas CV, Walker JD, Mukherjee R, Hebbar L. Time-dependent changes in matrix metalloproteinase activity and expression during the progression of congestive heart failure: relation to ventricular and myocyte function. Circ Res 1998;82:482–95. [DOI] [PubMed] [Google Scholar]
- 185.Shinbane JS, Wood MA, Jensen DN, Ellenbogen KA, Fitzpatrick AP, Scheinman MM. Tachycardia-induced cardiomyopathy: a review of animal models and clinical studies. J Am Coll Cardiol 1997;29:709–15. [DOI] [PubMed] [Google Scholar]
- 186.Riegger AJ, Liebau G. The renin-angiotensin-aldosterone system, antidiuretic hormone and sympathetic nerve activity in an experimental model of congestive heart failure in the dog. Clin Sci (Lond) 1982;62:465–9. [DOI] [PubMed] [Google Scholar]
- 187.Monnet E, Orton EC. A canine model of heart failure by intracoronary adriamycin injection: hemodynamic and energetic results. J Card Fail 1999;5:255–64. [DOI] [PubMed] [Google Scholar]
- 188.Toyoda Y, Okada M, Kashem MA. A canine model of dilated cardiomyopathy induced by repetitive intracoronary doxorubicin administration. J Thorac Cardiovasc Surg 1998;115:1367–73. [DOI] [PubMed] [Google Scholar]
- 189.Hyldebrandt JA, Siven E, Agger P et al. Effects of milrinone and epinephrine or dopamine on biventricular function and hemodynamics in an animal model with right ventricular failure after pulmonary artery banding. Am J Physiol Heart Circ Physiol 2015;309:H206–12. [DOI] [PubMed] [Google Scholar]
- 190.Aguero J, Ishikawa K, Hadri L et al. Characterization of right ventricular remodeling and failure in a chronic pulmonary hypertension model. Am J Physiol Heart Circ Physiol 2014;307:H1204–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.van Duin RWB, Stam K, Cai Z et al. Transition from post-capillary pulmonary hypertension to combined pre- and post-capillary pulmonary hypertension in swine: a key role for endothelin. J Physiol 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Aguero J, Ishikawa K, Fish KM et al. Combination proximal pulmonary artery coiling and distal embolization induces chronic elevations in pulmonary artery pressure in Swine. PLoS One 2015;10:e0124526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Pereda D, Garcia-Lunar I, Sierra F et al. Magnetic Resonance Characterization of Cardiac Adaptation and Myocardial Fibrosis in Pulmonary Hypertension Secondary to Systemic-To-Pulmonary Shunt. Circ Cardiovasc Imaging 2016;9. [DOI] [PubMed] [Google Scholar]
- 194.Sage E, Mercier O, Herve P et al. Right lung ischemia induces contralateral pulmonary vasculopathy in an animal model. J Thorac Cardiovasc Surg 2012;143:967–73. [DOI] [PubMed] [Google Scholar]