- Review
- Open access
- Published:
Advances in humanoid organoid-based research on inter-organ communications during cardiac organogenesis and cardiovascular diseases
Journal of Translational Medicine volume 23, Article number: 380 (2025)
Abstract
The intimate correlation between cardiovascular diseases and other organ pathologies, such as metabolic and kidney diseases, underscores the intricate interactions among these organs. Understanding inter-organ communications is crucial for developing more precise drugs and effective treatments for systemic diseases. While animal models have traditionally been pivotal in studying these interactions, human-induced pluripotent stem cells (hiPSCs) offer distinct advantages when constructing in vitro models. Beyond the conventional two-dimensional co-culture model, hiPSC-derived humanoid organoids have emerged as a substantial advancement, capable of replicating essential structural and functional attributes of internal organs in vitro. This breakthrough has spurred the development of multilineage organoids, assembloids, and organoids-on-a-chip technologies, which allow for enhanced physiological relevance. These technologies have shown great potential for mimicking coordinated organogenesis, exploring disease pathogenesis, and facilitating drug discovery. As the central organ of the cardiovascular system, the heart serves as the focal point of an extensively studied network of interactions. This review focuses on the advancements and challenges of hiPSC-derived humanoid organoids in studying interactions between the heart and other organs, presenting a comprehensive exploration of this cutting-edge approach in systemic disease research.
Introduction
The progression of cardiovascular diseases (CVDs) involves significant functional and structural alterations in vital organs and tissues [1]. Under stress and pathological conditions, the body regulates the balance of glomerular filtration rate and blood pressure through sympathetic stimulation, culminating in interaction relationships within the heart-brain-kidney axis [2]. The inflammatory response induced under pathological conditions plays a crucial role in inter-organ communication through cytokine secretion. This leads to interconnected communication between the heart and lungs [3], as well as between the heart and liver tissues [4]. Additionally, alterations in gastrointestinal microbiota [5] and neurologic damage in the brain [6, 7] have been reported to be strongly associated with the onset and progression of CVDs. Understanding the mechanisms of inter-organ communications is imperative for developing more precise drugs and effective treatments for CVDs and related organ pathologies.
Animal models are valuable tools for studying complex inter-organ communication, particularly in allergic and other non-infectious systemic diseases characterized by multifactorial effects [8]. As a significant relay organ, the spleen has also been shown to reduce infarct size in animal models by releasing cardioprotective factors during vagal activation [9]. Despite their effectiveness, animal models have some limitations, including significant interspecies differences in genetic expression and environmental factors, which hinder the replication of human conditions. Critically, patterns of gene expression regulation, inflammatory responses, and metabolic activity differ significantly between animal and human hearts [10]. To overcome these limitations and more realistically simulate physiological and pathological mechanisms, there is an urgent need for in vitro humanoid organoid or tissue models.
The discovery and development of human pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), has ushered in a new era of transformative research into the specific development of human organs and diseases [11]. Notably, the distinctive capability of hiPSCs to faithfully replicate a patient’s genetic background makes them an invaluable in vitro platform for studying the effects of critical cell-cell interactions (CCIs) and inter-organ communications in pathological situations, thereby enhancing the relevance of findings to patient-specific conditions [12]. hiPSCs can differentiate into specific organ-specific cells, allowing for the investigation of organ interactions through two-dimensional (2D) cell co-culture [13]. With the continuous advancements in biotechnology and tissue engineering, recent developments in three-dimensional (3D) organoid technologies, such as multilineage organoids, assembloids, and organoids-on-a-chip, which provide enhanced structural and functional fidelity, offer great potential for constructing functional humanoid organoids from hiPSCs. These advancements facilitate the study of inter-organ communications during organism development and disease progression [14]. Furthermore, emerging omics technologies such as single-cell and spatial transcriptomics enable more sophisticated analysis of these heterogeneous and complex tissues [15].
This review focuses on developing and applying hiPSC-derived 2D and 3D models for investigating heart-organ interactions under both physiological and pathological conditions. Additionally, it explores emerging omics techniques used in the study of inter-organ communications.
Application of 2D co-culture models in the study of CCIs
Understanding the cardiovascular system requires a multilevel and multifaceted approach that encompasses multiple organs and cell types. 2D co-culture models based on the hiPSCs can build bridges between different specific cells and investigate potential interaction effects [16, 17]. In the cardiovascular field, the current 2D co-culture model is mainly concerned with the interaction effects of cardiomyocytes with the neural-immune microenvironment, which are crucial for understanding cardiac function and pathology.
While the innervation of the autonomic nervous system in the heart is critical in the cardiac pathophysiological, the neurocardiac co-culture model provides an essential method for exploring the signaling effects of the heart in response to external stimuli, and for studying cardiac arrhythmias, neuro-cardiotoxicity, and safety assessment [18, 19]. The roles of both the sympathetic and parasympathetic nervous systems in the heart have been studied using the neurocardiac co-culture model. Yoh-Suke Mukouyama and colleagues developed a direct co-culture model of sympathetic neurons with cardiomyocytes, demonstrating that the presence of sympathetic neuron upregulated contractile function-related genes in cardiomyocytes [20], such as those encoding fine myofibrillar filaments and thick myofibrillar filaments, as well as genes related to voltage-dependent Na+, and inwardly rectifying K+ ion channels. Structurally, sympathetic neurons also promoted the formation of gap junctions in cardiomyocytes via connexin 43 (CX43), facilitating intercellular contact and allowing action potential propagation. To explore whether the parasympathetic nervous system can establish functional connections with target tissues, Nadja Zeltner et al. constructed a direct co-culture model of cardiomyocytes and parasympathetic neurons [21]. This model revealed a reduction in cardiomyocyte beating in response to acetylcholine receptor stimulation on parasympathetic neurons. Additionally, exposure to inflammatory factors such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), in combination with indirect co-culture in parasympathetic neuron medium, significantly restored reactive oxygen species (ROS) levels and beating variability in pathological cardiomyocytes, bringing them closer to the levels observed in healthy cardiomyocytes. These findings underscore the dual regulatory role of the nervous system in cardiac tissue, with the exact mechanisms still warranting further investigation.
The functional role of macrophages in the microenvironment of the infarcted heart, especially their interactions with cardiovascular cells, remains an area of ongoing investigation [22]. In response, the immune-cardiac co-culture model has emerged. hiPSC-derived allogeneic macrophages significantly improved cardiac function by injection into the infarcted myocardium of rats, mainly as a result of the exchange of damaged macrophages with healthy ones [23, 24]. Macrophages exchange sodium, potassium, and calcium ions through gap junction protein 43, either directly or indirectly [25]. In co-cultures of cardiac macrophages and iPSC-CMs, direct membrane contacts are formed, exerting electrophysiological modulation on the contractility of cardiomyocytes [26]. These findings suggest that the immune-cardiac co-culture model is a promising platform for studying ion channel diseases such as arrhythmias. By modulating macrophage states in inflammation, fibrosis, tissue regeneration, and repair, this model can also provide insight into therapeutic strategies for adult myocardial infarction [27, 28].
In addition to macrophages, other immune cells also play a significant role in maintaining cardiac homeostasis. By using the direct co-culture model, Kunal Sikder and colleagues investigated the mechanism of leukocyte-cardiomyocyte crosstalk [29]. They found that elevated levels of interleukin-1 beta (IL-1β) and TNF-α induce leukocyte recruitment to the injured tissues, where increased expression of alpha-smooth muscle actin (αSMA) and intercellular cell adhesion molecule-1 (ICAM-1) indicates leukocyte activation and migration. Furthermore, IL-1β and TNF-α enhance chemokine chemotaxis to the injured area via the danger-associated molecular patterns (DAMP) signaling pathway, disrupting cardiac homeostasis and contributing to the progression of chronic heart failure through cardiomyocyte loss.
Preadipocytes also secrete large amounts of pro-inflammatory cytokines [30]. Ichiro Morioka et al. explored the interaction between epicardial adipose tissue and myocardial tissue using an indirect co-cultured model [31]. They found that human adipose-derived dedifferentiated adipose cells secret various pro-inflammatory mediators, such as chemokine ligands 1 and 12, granulocyte colony-stimulating factor, IL-6 and IL-8, macrophage migration inhibitory factor This led to decreased cardiomyocyte function, as evidenced by reduced contraction frequency and increased oxygen consumption rates, contributing to a better understanding of the pathogenesis of epicardium-associated heart failure.
The heart and vascular network are the two most critical components of the cardiovascular system. Current reports highlight the concern of heart failure with preserved ejection fraction, particularly in women, suggesting that postmenopausal estrogen deficiency may be involved in the pathogenesis. Eiki Takimoto and colleagues explored the crosstalk between endothelial cells and cardiomyocytes using a direct co-culture model to uncover the mechanisms underlying estrogen deficiency-associated heart failure with preserved ejection fraction [32]. They demonstrated a pivotal role of the endothelial estrogen-myocardial cGMP (cyclic guanosine monophosphate) axis in angiogenic response and cardiac functional benefits, providing mechanistic insights into our understanding of sex differences in cGMP-related treatments for heart failure.
Despite the advantages of 2D culture models, such as ease of cultivation for high-throughput screening, they remain monotypic and lack the complex, self-contained matrix microenvironment inherent in 3D structures, which is essential for a nuanced interpretation of CCIs [33]. Meanwhile, current 2D cell-based in vitro disease models are limited in generalizing inter-organ communications [34]. Recent advancements in emerging technologies, including multilineage organoids, assembloids, and organoids-on-a-chip, have significantly improved our understanding of complex biological interactions, such as inter-organ communications.
Humanoid organoid models for studying inter-organ communications
Organogenesis is a coordinated process involving interactions among different embryonic tissues. 3D culture allows for the generation of in vitro tissues with organ functions similar to primary tissues [35]. Compared to the 2D cellular level, 3D tissue models are more effective in eliciting cellular fates resembling those observed in vivo during specific processes [36]. These organoids can effectively reproduce the complex structure of living organs and inherit the genetic background of the donor, making them invaluable tools for disease modeling, unravelling pathophysiological mechanisms, and elucidating developmental biology [37]. Research on cardiovascular organoids has advanced into various areas, including myocardium, blood vessels, and valves, providing numerous models for studying CVDs [38].
Combined with tissue engineering techniques, organoids-on-a-chip based on microfluidic devices have been developed [39]. It is now possible to assemble more complex tissue-assembled bionic organoids, which provide near-realistic models for studying human tissues and organs, as well as for simulating pathological processes [40]. Humanoid organoid models facilitate macroscopic and microscopic analyses of intercellular crosstalk, resulting in multi-organ models that effectively simulate complex tissue-tissue and inter-organ communications, making them suitable for organ development, toxicity, and efficacy [41]. This section describes the three main humanoid organoid models for studying inter-organ communications.
hiPSC-derived multilineage organoids
Most cardiac organoid studies have focused on individual organ genealogies or specific signaling pathways [42]. Although they show great potential in recapitulating the physiology of the human heart, they fail to capture the complex crosstalk between the heart and endodermal or ectodermal organs, limiting our understanding of integrated organ development [43]. Consequently, we still lack a comprehensive understanding of the development of multi-organ genealogies. Currently, organoids can self-assemble into ordered patterns and structures and generate multilineage organoids, a process guided by hiPSC aggregates and bi-directional Wnt signaling, which is crucial for mimicking developmental processes [44]. Moreover, recent reports indicate that hiPSC-derived multilineage organoids composed of cardiac and other tissues can reflect the structural and early developmental patterns of the embryonic heart, providing insights into the co-developmental relationship of cardiac development with foregut endoderm and ectoderm [45, 46]. Retaining cellular complexity, these organoids reveal critical structural features of the human heart and gut development.
Multilineage organoids provide an unprecedented opportunity for understanding organ development and shedding light on how the endoderm and heart cooperate to guide morphogenesis, patterning, and maturation. Specifically, these organoids recapitulate key morphogenetic events that occur primarily after heart tube formation and preserve these tissues and functions for up to 100 days [47]. Furthermore, multilineage organoids provide a microenvironment that permits the formation of intestinal tissue while promoting the maturation and stabilization of cardiomyocytes. Compared to animal models, multilineage organs provide a more direct view of tissue crosstalk mechanisms. As such, they represent a promising avenue for generating more physiologically relevant in vitro models of human development.
However, the reproducibility of multilineage organoids from each batch is a challenge due to the large size, high cell density, and inherent heterogeneity of multilineage organs. Additionally, the dynamic and complex processes within organoids necessitate collecting different batches of organoids independently, making the process time-consuming. The experimental variability of multilineage organoids, primarily stemming from the lack of external guidance for multilineage organoids and their reliance on self-organized structural reconstruction, results in unclear and dynamically unstable organoid compositions as they mature. The developmental patterns of different lineage vary across organs, highlighting the need for further clarification of the intersections between the developmental trajectories of multilineage organoids. Moreover, multilineage organoids still fall short of fully replicating the structure of mature organs, lacking the microenvironment and critical cellular interactions within the tissues. To address these limitations, efforts must focus on constructing organoids that better emulate the human microenvironment.
Assembloids
The sophistication of organoids is continually increasing to reflect the complexity of the human body’s multiple tissues. However, the cellular interaction network of multilineage organoids is not sufficient to form complex tissue microenvironments and lacks the required spatial organization [48, 49]. To address this limitation, assembloids composed of multiple organoids have been developed [50, 51]. Developed by Sergiu Paca’s group, assembloids represent an advanced extension of the 3D multicellular system [52]. Assembloids are created by combining two or more distinctly patterned organoids, integrating additional cell or tissue types. Assembloids are invaluable tools for modeling complex cellular integration within and between tissues, providing insights into the pathophysiology of various diseases [53].
Due to the lack of innervation and vascular network, cardiac organoids are unable to fully replicate the in vivo environment required for organ development, integrity, and functional coupling. To model organ complexity and heterogeneity, cardiac assembloids consisting of atria, atrioventricular canal cardiomyocytes, and ventricular globules have been developed to mimic the atrial-ventricular interface [54]. These models are particularly useful for studying complex cardiac conduction disorders, such as atrioventricular block, by reproducing the electrical propagation patterns seen in the heart. Moreover, cardiac organoids can be innervated with sympathetic neurons to construct neuro-cardiac assembloids, facilitating the modeling of inter-organ communications [55]. Anna F Rockel et al. developed a neuro-mesoderm assembly model, in which the co-developing of mesodermal and neuroectodermal tissues and the formation of a vascular plexus along with the peripheral nervous system was observed, allowing for the study of crosstalk between neuroectoderm and mesoderm, and between peripheral neurons/neuroblasts and endothelial cells [56].
Our group developed a methodology for generating vascularized and chamber-like cardiac organoids by enveloping hiPSC-derived aggregates destined for vascular fate with the differentiated cardiomyocytes. This configuration allows vascular-committed cells to migrate outward in response to a vascular endothelial growth factor (VEGF) gradient, thus vascularizing the peripheral myocardium [57]. This model facilitated the discovery of interactions between endothelial cells and cardiomyocytes through the PI3K-AKT signaling pathway, extracellular matrix (ECM) receptor interactions, and Ras signaling pathway. Notably, a cluster of neurocytes was identified in these cardiac assembloids, providing a unique platform for studying neuro-cardiac interactions in the future.
In order to investigate the critical cardio-pulmonary mutual interaction during human embryogenesis, Ng et al. developed a cardio-pulmonary assembly model by the 3D suspension culture of co-induced cardiac and pulmonary progenitors from hiPSCs [58]. While the cardio-pulmonary assembly is initially arranged in the pulmonary-centered, concentric manner, the heart and lung compartments gradually move away from each other, eventually separating from each other, mimicking the distinct tissue development seen in the body. Therefore, these assembloids provide a powerful tool for observing organ interactions, though further investigation is needed to understand the specific regulatory mechanism involved.
In addition to cardiac assembloids, brain organoid assembloids containing multiple brain regions have been developed for elucidating and treating neurodevelopmental disorders [59, 60]. Based on these assembloids, studies have explored interactions between the nervous and immune systems [61], nerve-muscle organoids [62], and functional vascular brain assembloids consisting of human cortical organoids and the vascular system [63]. With advances in 3D bioprinting technology, which allows for precise control over the size, shape, structure, and organization of the printed tissues, more robust and scalable assembloid systems can be developed [61]. Despite these advancements, a human heart-brain assembloid has yet to be developed or tested, particularly using iPSCs derived from patients with pathological dysfunctions in both heart and brain tissues, such as those seen in Dravet syndrome caused by haploinsufficiency of the sodium voltage-gated channel alpha subunit 1 (SCN1A) gene [64]. These models can provide valuable insight into the role of target genes in the heart-brain axis.
The complexity of assembloids depends on two key factors, the generation of specific cell types and their diversity, which result in significant heterogeneity in assembloid components [65]. Moreover, it’s important to note that the culture standards for assembloids have not yet been fully standardized. Assembloids lack precise techniques to quantify various aspects of pathophysiology. More refined and complex microfluidic technologies are needed to serve as a platform for the mass production of subsequent models. Combining these advancements with emerging omics technology will enhance the relevance of experimental results to personalized clinical patients, significantly improving the feasibility of clinical translation.
Organoids-on-a-chip
To simulate more complex microenvironments of multi-organ systems, current 3D tissue construction techniques combined with tissue engineering enable the creation of environments more compatible with human systems [66]. Organoids-on-a-chip refers to the integration of multiple organoid systems in a co-culture system, achieved by linking various organoid models within a microfluidic device [67]. This setup facilitates the co-culture of different types of organoids in a regionalized microenvironment. Specially, hiPSC-derived organoids-on-a-chip technology offers the advantage of incorporating the complete polygenic disease background of the patient [68]. This approach not only avoids ethical concerns but also leverages CRISPR/Cas9 technology to modify gene interactions, allowing for the modeling of complex diseases.
As a research platform that incorporates cellular and organoid models, organoids-on-a-chip effectively recaptures the communication between connected organs [69]. Organs-on-a-chip supports inter-organ communications by precisely controlling chemical, physical, and cellular micro-environments. This modular platform could effectively mimic barrier effects and mechanical interactions between different organ tissues [70]. Additionally, organoids-on-a-chip demonstrates reproducibility, including tumor structure, heterogeneity, function, and genetic stability in culture [71]. These systems also allow real-time monitoring of cellular responses to various stimuli, revealing disease mechanisms and their role in signaling pathways.
Organoids-on-a-chip has been used to study the interactions of multiple organ axis. For example, studies have focused on the interactions of intestinal tissue with other organs. This includes investigating phenomena such as the barrier effects of the gut-brain axis, the metabolic dynamics of the gut-hepatic axis, drug absorption, and toxicity assessment through the intestinal-renal axis, the telecommunications effects of the gut-lung axis, and the immune interactions of the enteropancreatic axis [72]. Furthermore, the complex network of biochemical signals and mechanical forces in the bone-organ axis has been applied to explain mechanisms related to bone disease, drug screening, and pharmacokinetic analysis. These platforms have also been utilized to model complex physiological and pathological responses [73]. Based on these applications, the interactions between the heart and other organs in organoids-on-a-chip will be discussed in the following sections.
Despite the advancements, the application of microfluidic organoid chips faces three main challenges. First, the interactions between the microfluidic device environment and the cells, as well as between the cells within the device, remain underexplored. Emerging artificial intelligence (AI) techniques, such as large language models and AI-based analysis, may provide potential solutions for these challenges. Secondly, organoid culture processes lack a complex cellular microenvironment and rely on animal-derived matrices, such as substrates, whose composition is not well defined. Long-term culture and viability remain potential challenges. Lastly, the lack of standardized culture protocols is a significant limitation. Variations in patient samples, culture methods, media choices, and the absence of standards for organoid maturity all impact the consistency and application of organoids-on-a-chip models in clinical translational research.
Application of humanoid organoid models
Humanoid organoid models have great potential for simulating organogenesis, disease modeling, and drug screening at the multi-organ systems level, with the ultimate goal of playing a pivotal role in personalized medicine research. In order to understand the mechanisms governing interactions between the heart and multiple other organs in the organism, more targeted and effective therapeutic strategies must be developed. Based on the above models, communications between the heart and other organs have emerged as a significant field of study, which will be primarily discussed in the following context (Fig. 1), and summarized in Table 1.
Humanoid organoid-based models for studying heart-organ interactions. The development of hiPSC-derived multilineage organoid offers a platform for investigating germ layer interactions during cardiac organogenesis. Two primary models including assembloids and organoids-on-a-chip, have been created to model heart-organ interactions under both physiological and pathological conditions, advancing our understanding of underlying mechanisms and supporting drug screening efforts. Created in BioRender. Ni, B. (2024) BioRender.com/p02w371
Heart-organ axis-based organogenesis
Endodermal, mesodermal, and ectodermal structures can differentiate into specific tissues and organs. Some findings indicate defects in the thyroid gland and heart when the interplay between the germ layers is disturbed [74], underscoring the importance of germ-layer communications. Recent single-cell transcriptomics is elucidating the biochemical and molecular mechanisms of paracrine signaling in endoderm-mesoderm communications [75]. The hepatobiliary-pancreatic axis, consisting of designated endoderm and mesoderm, is designed to mimic the genesis and development of dynamic organs in complex environments, providing opportunities for studying multi-organ diseases in humanoid organoid models [76]. Additionally, it has been demonstrated that inter-organ, cross-lineage communications occur between the co-developing heart and the surrounding endoderm [77]. Although human cardiac organoids show great potential in recapitulating the physiology of the human heart, they often fail to capture the complex crosstalk between the heart and the endodermal organs. Thus, a comprehensive understanding of multi-organ lineages remains elusive. Here, we will expand the application of 3D organoid models in studying interactions between the heart and other endodermal organs.
Heart-foregut axis
A human multilineage pro-epicardium/foregut organoid has been developed to recapitulate the co-emergence of the anterior epicardium, septal mesenchyme, and liver buds. This model investigates its supporting role in epicardium/myocardium development [78]. It primarily involves the co-culture of these multilineage organoids with cardiomyocyte aggregates, forming self-organizing cardiac organoids in which the epicardial-like layer surrounds the myocardial-like tissue. This study offers novel interpretations that advance our understanding of myocardial-foregut interactions during cardiac organogenesis in healthy or diseased environments.
In a related study, Robert Zweigerdt and colleagues encapsulated hiPSC-derived aggregates in Matrigel to simulate early cardiac development, which also forms foregut endodermal tissue, under the influence of Wnt activators and inhibitors [79]. Since the interplay between cardiac mesoderm and foregut endoderm is required for proper myocardial- and endocardial patterning, this model offers a valuable tool for studying in vitro congenital cardiac developmental defects caused by genetic defects. Notably, they observed reduced cardiomyocyte adhesion and hypertrophy in the presence of NKX2.5 mutations, a phenotype similar to the cardiac malformation seen in NKX2.5-knockdown mice. These findings offer new perspectives for understanding developmental and genetic disorders in cardiac biology.
Heart-intestinal axis
Todd C. McDevitt and colleagues developed a multilineage organoid model that recapitulates embryonic heart and gut development [47]. In this model, aggregates consisting of hiPSC-derived mesendoderm progenitors are directed toward a cardiac differentiation fate with ascorbic acid supplementation. The long-term culture of these multilineage organoids successfully mimics the development of embryonic heart and gut. This model suggests that endoderm-derived cells produce paracrine signals and mechanical forces that support cardiac maturation. Interactions between the heart and intestinal tissues give rise to a more structurally and functionally mature cardiomyocyte population and suggest the induction of an atrial/lymph node differentiation fate.
The multilineage organoid model provides a more direct approach to studying genetic disorders that affect the heart and gut, such as chronic atrial and intestinal rhythm disorders [80]. Furthermore, these multilineage organoids offer a unique opportunity to model tissue morphogenesis or multi-organ complex diseases in vitro, and enable the examination of multi-tissue/organ interactions in the context of embryonic organoid lineage development, physiological maturation, and disease conditions.
Heart-pulmonary axis
As the two major organs in the thoracic cavity, the extensive crosstalk between the developing heart and lungs is crucial for their joint morphological stability and functional maturation. Extensive cardio-pulmonary interactions during organogenesis have been well demonstrated in mouse models [81]. The involvement of pulmonary circulation in cardiac specialization suggests that T-box transcription factor 5, a transcription factor required for cardiac segregation in the second heart field [82], directly drives the cardiac morphogenetic gene regulatory network. Additionally, the conserved retinoic acid-Hedgehog-Wnt signaling cascade coordinates cardiopulmonary development [83]. These pathways initiate a signaling axis of mesoderm-endoderm interactions, providing a molecular basis for the co-evolution of lung and heart structures.
Recent advancements in modeling cardiopulmonary interactions within concentric 3D suspension cultures have further enhanced our understanding [84, 85]. Specifically, Ng WH and colleagues generated heart-lung assembloids by clustering cardiac progenitors and lung progenitors, which were derived from hiPSCs by regulating the Wnt and Nodal signaling pathways, respectively. Their findings demonstrated that the presence of heart tissue accelerated the maturation of lung progenitor cells. Over time, the heart and lung compartments gradually separated, becoming independent. Notably, the activation of exogenous Wnt through GSK-3β inhibition effectively slowed this heart-lung separation, offering valuable insights into the cellular and molecular mechanisms underlying cardiopulmonary co-development and tissue boundary fusion. Although the exact mechanisms remain unclear, this model holds significant clinical potential, particularly in understanding congenital cardiac and pulmonary diseases, such as Diaphragmatic Hernia [86]. Meanwhile, mapping the temporal and spatial relationships between cardiopulmonary and cardiac lineages is expected to provide fundamental insights into the crosstalk essential for organ development.
Heart-organ axis-based cardiovascular disease modeling and drug screening
In addition to studying the intercellular and inter-organ communications during cardiac development, multilineage organoids also offer a platform to simulate the developmental processes of conditions such as innate cardiac dysplasia [87, 88]. Considering the complex microenvironment of the organism, combining existing assembloids with microfluidic technologies enables the establishment of multi-organ disease models. This approach is beneficial for studying mechanisms related to multi-organ system diseases and screening for more effective drugs with fewer side effects, ultimately improving patients’ quality of life and survival rates.
Heart-kidney axis
Heart and kidney diseases are closely linked due to their interdependent functions. Cardiorenal syndrome, which involves nephropathy and cardiac disease, represents a common physiological and pathological condition affecting these organs [89]. Dysfunction in the kidneys, through interactions with the circulatory system, usually leads to increased circulating volume and, ultimately, heart failure [90]. However, the complex mechanisms of dysfunction between the cardiac and kidney organoids in multi-organ diseases remain poorly understood [91]. While single-organ models such as cardiac and renal organoids have significantly advanced biomedical research, they cannot capture the dynamic interactions between these organs.
The in vitro models that combine hiPSC-derived heart and kidney organs on a microfluidic chip to form a heart-kidney axis are proving valuable for the study of heart-kidney interactions [92]. These chips typically feature separate channels connecting distinct chambers, with microfluidic channels ensuring the structural and functional integrity of each organ under both static and dynamic conditions. Using this model, the coculture of renal organoids has been shown to reduce the contraction amplitude of cardiac organoids, a phenomenon attributed to nutrient consumption, such as glucose, by the renal organoids [92]. As an in vitro microfluidic platform for the cardiorenal axis, this model enables the investigation of the heart’s response to stress or injury and allows for the assessment of how cardiac injury impacts renal tissue, including alterations in renal blood flow, inflammatory responses, and fibrosis [93]. Moreover, from a clinical perspective, this model can be used to investigate the therapeutic effects of cardiac drugs on cardiac tissue and evaluate their possible nephrotoxic effects.
The proximal tubules in the renal unit interact with the vascularization of the cardiac circulatory system through both direct actions (secretion of factors) and indirect actions (glucose and nutrient uptake). However, since cardiac organoids typically lack vascularized structures, combining microarray technology with assembloids is essential to create more complex bionic organoid structures. This integration would better mimic the glomerular and tubule-cardiac circulatory system crosstalk, providing a more accurate representation of the physiological interactions between the heart and kidneys.
Heart-liver axis
Drug-induced hepatic and cardiac side effects are primary challenges leading to drug failures in clinical trials [94]. A multi-organ microarray system developed by Jianhua Qin and colleagues successfully maintained the tissue-specific functions of both cardiac and liver organoids in a co-culture setup [95]. This system was used to assess the liver metabolism in drug-induced cardiotoxicity assays. They found that exposure of Clomipramine, a widely used antidepressant drug, to liver organoids on chip led to impaired cardiac function, significant cardiomyocyte death, and reduced release of calcium fluxes [95]. This is consistent with the side effects observed in clinical patients treated with Chlorpromazine, leading to the conclusion that Chlorpromazine-induced cardiotoxicity is dependent on hepatic metabolism. A multi-organ system of functional, non-invasive recordings has also been devised to study crosstalk between the heart and liver. This model simulates the hydrodynamic impact of drugs in the cardiac and hepatic systems by generating gravity-driven flow through a rocking arm platform, thereby predicting drug metabolism concentration changes more accurately [96].
The liver can metabolize certain drugs, either inactivating or activating their effects. Metabolites produced in the liver, such as Adriamycin, can have toxic effects on the heart, making it essential to characterize liver metabolism when studying the cardiac effects of drugs. Recent articles have summarized recently developed liver and cardiac microarray systems for drug toxicity testing [97]. These systems have shown the potential to predict in vitro action mechanisms and mimic patient-specific differences, suggesting that combining microarray technology with organoid technology is an effective model for in vitro drug screening.
Heart-neuro axis
The heart’s innervation is predominantly regulated by the autonomic nervous system, comprising the sympathetic and parasympathetic branches [98]. Changes in neuronal activity significantly affect the structure and function of cardiomyocytes [99]. In turn, heartbeat-induced pressure pulsations have been shown to modulate neuronal activity via mechanosensitive ion channels [100, 101]. These findings illuminate the functional role of coupling between neurons and the heart, highlighting the coordinated responses that may occur during specific activation states. In the neuro-cardiac axis, which mainly involves interactions between the exogenous and endogenous cardiac nervous system, it is essential to simulate changes in the autonomic nervous system at the in vitro level in both normal and pathological states. This approach allows for the study of diseases such as cardiac arrhythmias at a more complex level, focusing on neural regulation rather than direct interventions in the cardiomyocytes [102, 103].
Leveraging the rapid advancements in tissue engineering and organoid technologies, researchers have applied neuro-cardiac organoids on microarrays to demonstrate functional synapses and the contractile coupling properties of cardiac calcium dynamics induced by neuronal stimulation. Specifically, Albano C. Meli and colleagues developed an organ-on-a-chip system in which neuronal cells connected to hiPSC-CMs, forming functional synapses between neurons and cardiomyocytes [99]. From a clinical translational point of view, their models were also tested with acetylcholine analogs: Atropine and Carbachol. They demonstrated that Atropine did not substantially affect the electrical signals of the heart, whereas Carbachol increased the amplitude of calcium transients in cardiomyocytes, potentially suggesting concomitant cardiotoxic effects of Carbachol.
Models are available that incorporate novel engineered materials and developmental features to articulate the neural-cardiac axis [104]. This will further increase the complexity and accuracy of cardiac organoid models in response to autonomic nervous system innervation. Ultimately, this integration contributes to the development of more effective therapeutic strategies, which can reveal the specific effects of the autonomic nervous system on cardiac function.
Multi-organ interactions
Organs communicate with each other primarily through specific tissue boundaries, unique inter-organ barrier effects, and crosstalk facilitated by vascular flow. Diseases affecting multiple organ systems pose significant challenges in simulation, and there remains a lack of in vitro models that can accurately represent the interactions of human tissues across various organs, particularly humanoid disease models.
A three-tissue organ-on-a-chip system, comprising liver, heart, and lung organoids, has been developed to study inter-organ responses to drugs and other compounds [105]. In this system, bioengineered tissue organoids and tissue constructs are integrated within a closed circulatory perfusion system, replicating the interactive nature of the human body. An unanticipated side effect of Bleomycin, cessation of cardiac organoid beating, was detected in the three-tissue organ-on-a-chip system. This side effect was not merely due to Bleomycin but potentially caused by a secondary factor produced by one of the other tissues in the platform, since this effect was not observed in a cardiac-only system, highlighting the value of multi-organ interaction models [105].
Recently, Aleksander Skardal’s lab expanded the integrated system to incorporate six humanized constructs, including liver, cardiac, lung, endothelium, brain, and testes organoids [106]. Drug toxicity assessment in the six-organoid system demonstrated that the anticancer drug Ifosfamide exhibits significant neurotoxicity when liver organoids are present. These findings underscore the importance of liver-associated metabolism in the neurotoxic effects of circulating Ifosfamide.
Given that blood flow is intricately linked to the development of CVDs, a multi-organ model featuring the vascular barrier was also created. This model incorporates mature myocardial, liver, bone, and skin tissues within a multi-organ microarray to detect inter-tissue crosstalk effects. This system, using fluorescent markers secreted by different tissues, unexpectedly revealed the involvement of immune cells in the vascular barrier [107, 108]. Further studies using inter-organ tissue microarrays hold promise for more accurately modeling multi-systemic diseases such as CVDs, for identifying early biomarkers for drug toxicity [109].
In summary, multi-organ microarray systems, particularly those incorporating the heart, are essential for evaluating the safety and efficacy of drugs that have direct or indirect effects on cardiac function. As AI-driven big data technologies and advanced histology techniques continue to evolve, these systems are poised to enhance the success rate of new drug development.
The maturation of cardiomyocytes in hiPSC-derived cardiac models
Although hiPSC-derived cardiac organoids have emerged as a promising tool for studying cardiac physiology and pathology, and inter-organ communications, cardiomyocytes in these organoids exhibit several immature and fetal-like features that limit their functional resemblance to adult cardiomyocytes. Understanding these differences is crucial for advancing their application in disease modeling and therapeutic interventions.
Immature features of hiPSC-derived cardiomyocytes
Morphology
Mature human cardiomyocytes typically exhibit a unique and well-defined rod-like structure, with a diameter of 15–30 μm and a length of 100–150 μm, which is essential for normal cardiac function [110]. In contrast, hiPSC-CMs are smaller, with diameters ranging from 5 to 10 μm, and often display irregular, round shapes [111]. Meanwhile, hiPSC-CMs exhibit irregular sarcomeres and T-tubules, as well as less abundant mitochondria and gap junctions, leading to a disorganized contraction. Altogether, these morphological differences influence their electrophysiological and contractile properties, thereby limiting their utility in disease modeling.
Electrophysiology
The electrophysiological properties of cardiomyocytes are vital for normal heart function and in response to pathological states. hiPSC-CMs often exhibit an imbalance between sodium-calcium exchanger and L-type Ca2+ channel-mediated inward currents and potassium ion efflux, which affects cardiac electrophysiologic activity and contractile function [112]. Unlike the adult ventricular cardiomyocytes, which are electrically quiescent until triggered by a neighboring cell, the immature hiPSC-CMs exhibit also autorhythmicity. This characteristic is due to the high levels of hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) in the plasma membrane and the spontaneous leak of calcium from the sarcoplasmic reticulum [113].
Metabolism
During maturation, cardiomyocytes transition from glycolysis to fatty acid β-oxidation as their primary energy source, accompanied by increased mitochondrial density and more developed cristae structures [114]. In contrast, immature cardiomyocytes predominantly rely on glycolysis, with weaker mitochondrial oxidative phosphorylation and lower ATP production, for energy supply [115]. This metabolic immaturity may lead to biases in drug toxicity assessments and hinder effective clinical translation.
Excitation-contraction coupling property
Excitation-contraction coupling properties are fundamental to cardiomyocytes and are essential for other electrophysiological and mechanical cellular properties. Mature cardiomyocytes utilize T-tubular junctions for synchronized calcium release during action potential conduction, maintain high sarcoplasmic reticulum calcium content, and exhibit tight coupling of L-type calcium channels to ryanodine receptors to sustain Ca2+ kinetic equilibrium [116]. hiPSC-CMs, however, possess immature T-tubular structures, affecting intracellular calcium release and resulting in immature calcium transients and conditional dependence on calcium cycling. These immaturity challenges the accurate replication of mechanical characteristics observed in the adult heart.
Methods and challenges in maturing hiPSC-derived cardiomyocytes
To enhance the maturity of hiPSC-CMs, a variety of strategies have been proposed to better mimic the physiological environment or modulate key signaling pathways. These methods primarily focus on long-term culture, biophysical stimulation, cell co-culture, and 3D culture. While promising, each of these strategies presents its own challenges and limitations that must be addressed to optimize their application in disease modeling, drug screening and regenerative medicine.
Long-term culture
Long-term culture is a well-established approach to promote the maturation of hiPSC-CMs by closely mimicking the time course of cardiac development. After more than 60 days of culture, hiPSC-CMs show improved myofilament organization, and undergo a metabolic shift, with increased mitochondrial number, mature cristae structures, and more efficient ATP production. A significant improvement in contractility and calcium-handling capacity will be also observed in hiPSC-CMs after a long-term culture [113, 117, 118].
Biophysical stimulation
Biophysical stimulation, including mechanical and electrical signals, is another promising strategy to drive the maturation of hiPSC-CMs by mimicking the dynamic mechanical and electrophysiological microenvironment of adult cardiomyocytes. Adult cardiomyocytes are subjected to cyclic stretch and fluid shear forces during cardiac systole and diastole, which are essential for regulating gene expression and functional maturation [119]. By applying cyclic stretch to hiPSC-CMs, their myotome length increases, and the alignment of transverse striations is enhanced [120]. Meanwhile, electrical stimulation promotes the maturation of action potentials and improves calcium transient synchronization in hiPSC-CMs [121].
Co-culture and 3D culture
Co-culture and 3D culture are advanced strategies designed to more accurately replicate the cardiac microenvironment by fostering physical, chemical, and intercellular interactions that promote maturation. Co-culture with other cell types, such as cardiac fibroblasts, endothelial cells, and mesenchymal stem cells, provides paracrine signaling and direct cell-to-cell interactions that support maturation [122,123,124]. For instance, co-culturing hiPSC-CMs with cardiac fibroblasts promotes functional coupling, reduces spontaneous beating frequency, and prolongs action potential duration [122]. 3D culture systems, through mimicking the cell component and mechanical properties of myocardial tissues, could promote the development of formation of T-tubule systems and accelerate the maturation process. Recent advancements in 3D co-culture systems show great promise in enhancing maturation and reducing the time required for functional development [122, 125, 126].
Despite significant progress, existing methods to promote the maturation of hiPSC-CMs still face limitations, particularly in replicating adult myocardial tissue’s contractility, metabolic capacity, and functional maturation. Long-term culture is time-consuming and complex, with the potential for cell aging or functional degradation [127]. While co-culture and 3D culture systems offer more in vivo-like representations, challenges persist in optimizing cell ratios (e.g., avoiding fibroblast-induced fibrosis) and managing the heterogeneity that may interfere with drug screening or signaling studies [128]. 3D culture further faces issues such as technical complexity in vascularization and nutrient/oxygen gradients leading to cell necrosis [57, 129]. Additionally, the lack of standardized stimulation parameters and the need for scalable bioreactors complicate the process, with overstimulation potentially causing cellular damage and hindering effective maturation [123]. Variations in cell lines or culture conditions can also lead to inconsistent maturation, affecting disease modeling accuracy.
Application limitations of immature cardiomyocytes
Given the ongoing challenges in achieving adult-like hiPSC-CMs, their limitations must be carefully considered when applying these hiPSC-derived cardiac models, as outlined in the following sections.
Inadequate functional simulation
Immature hiPSC-CMs exhibit weak and uncoordinated contractility, which limits their ability to replicate the synchronized contractile behavior of adult myocardium. This immaturity reduces the sensitivity of models in assessing myocardial dysfunction, particularly in diseases like heart failure [130]. Additionally, the electrophysiological properties of immature hiPSC-CMs differ significantly from those of adult cardiomyocytes, with notable discrepancies in action potential morphology and ion channel expression [131]. These differences impair the ability of hiPSC-CMs to coordinate electrophysiological activity with cells from other organ systems, such as neurons, thereby affecting the accuracy of inter-organ signaling simulations [132].
Bias in disease modeling
In models of inherited cardiomyopathies, such as hypertrophic cardiomyopathy (HCM), the immaturity of hiPSC-CMs may obscure the phenotypic expression of disease-causing mutations. For instance, mutations in sarcomeric proteins that lead to severe dysfunction in mature tissues may induce minor contractile abnormalities in immature cells [133]. Furthermore, variations in cell lines or culture conditions can lead to differences in maturation, exacerbating biases in disease modeling [127]. The paracrine signaling profile of hiPSC-CMs also diverges from that of adult cardiomyocytes, which can distort disease-related signaling pathways. For example, cytokines like IL-6, secreted by hiPSC-CMs, may induce aberrant inflammatory responses in co-cultured cells, confounding disease modeling and therapeutic testing [134].
Biased prediction of drug responses
The metabolic and electrophysiological immaturity of hiPSC-CMs can result in inaccurate predictions of drug toxicity. For example, the cardiotoxicity of certain chemotherapeutic agents is linked to mitochondrial function, but the glycolysis-dependent metabolism of immature hiPSC-CMs may lead to false-negative results [124]. In models of diseases like HCM, abnormal contraction patterns may not fully reflect the clinical condition of patients, thereby impairing the accuracy of drug testing [135]. Furthermore, metabolic differences between hiPSC-CMs and adult cardiomyocytes can limit the relevance of these models in studying the heart’s interaction with other metabolic organs (e.g., liver), which may obscure the effects of drug metabolism on cardiac function [136].
In summary, the immature characteristics of hiPSC-CMs impact the performance, reliability, and predictive accuracy of hiPSC-derived cardiac models in modeling cardiac physiology and pathology. To improve the maturity and application value of these models, multifactorial optimizations are necessary. An integrated approach, combining biomaterials, mechanical stimulation, metabolic modulation, and AI-assisted optimization, may help overcome these challenges and facilitate the effective application of hiPSC-CMs in precision medicine and translational research.
Application opportunities of immature cardiomyocytes
Despite their structural and functional immaturity, the unique features of hiPSC-CMs may not be a critical limitation in some applications and can even offer distinct advantages in certain application contexts. The following sections discuss how these immature characteristics can be effectively leveraged for specific applications.
Developmental biology and abnormality studies
The molecular and metabolic characteristics of hiPSC-CMs closely resemble those of fetal cardiomyocytes, making them ideal for constructing models of early cardiac development and associated developmental abnormalities. For instance, hiPSC-CMs have been utilized to investigate the pathogenesis of cardiac septal defects caused by embryonic GATA4 mutations [137]. Additionally, cardiac organoid models combined with machine learning have been used to study ventricular developmental defects caused by NKX2-5 deficiency [138]. Researchers have also successfully modeled cardiac outflow tract anomalies associated with DiGeorge syndrome, shedding light on the role of T-box transcription factor 1 in developing the cardiac conotruncal arterial trunks [139].
Modeling specific pathological features
Immature hiPSC-CMs are particularly valuable for modeling the early pathological features of specific cardiac diseases, such as inherited arrhythmias and cardiomyopathies, which are more easily triggered and observed in these cells [140]. For example, exposing hiPSC-CMs harboring a ryanodine receptor 2 (RYR2) mutation to β-adrenergic stimulation recapitulated the calcium spark abnormalities and delayed afterdepolarizations observed in patients with catecholaminergic polymorphic ventricular tachycardia, highlighting their propensity for arrhythmias [141].
Modeling non-myocyte-associated cardiac diseases
Non-myocyte cell populations in the heart, including fibroblasts, vascular cells, autonomic neurons, and immune cells, also play crucial roles in cardiac homeostasis and disease. For example, microvascular complications and neurocardiac dysfunction are commonly reported in patients with Type 2 Diabetes Mellitus, suggesting the involvement of non-myocyte cells in the disease process [142]. Cardiac organoids, which typically incorporate hiPSC-CMs along with additional cell types, offer a promising platform for studying the pathological mechanisms associated with these non-myocyte cells. Meanwhile, certain drugs may exert their effects on non-myocyte cell types. Recent studies have shed light on the potential mechanism of SLGT2 (sodium-glucose co-transporter-2) inhibitor-mediated cardioprotection, which may involve direct or indirect effects on the cardiac non-myocyte populations or even extra-cardiac tissues [142]. Therefore, despite the immaturity of hiPSC-CMs, hiPSC-derived cardiac organoids remain a valuable model for investigating the non-myocyte-related cardiac response to drugs or signals from other organs.
Drug screening and toxicity testing
Immature cardiomyocytes are more sensitive to drug-induced effects, making them valuable tools for detecting early toxicity or efficacy signals that may be missed in mature cardiomyocytes. For instance, screening for cardiotoxicity of the antiviral drug Raltegravir using hiPSC-CMs revealed a prolongation of the action potential duration, suggesting a potential arrhythmogenic risk [143]. These cells are also suitable for modeling hypersensitivity in pediatric hearts to drugs, such as the early cardiotoxicity of the chemotherapeutic agent Adriamycin [144].
In summary, rather than being a limitation, the immaturity of hiPSC-CMs offers unique advantages in modeling heart development during early stages and constructing sensitive disease models. By precisely regulating their maturation, the application potential of hiPSC-CMs in personalized medicine and translational research can be further expanded.
Omics tools for the integrative study of inter-organ communications
Studying inter-organ communications is essential for understanding complex physiological and pathological processes. Thus, comprehensive high-throughput techniques are indispensable for the integrative analysis of these interactions. Single-cell and spatial transcriptomics are current cutting-edge technologies offering detailed descriptions of physiological and chemical responses within tissues [145, 146]. These techniques enable the identification of biomechanical cues that facilitate cross-lineage interactions and predict signaling networks for tissue interactions [147]. By applying these techniques to the hiPSC-derived inter-organ interaction models, researchers can better understand the complex processes regulating multi-organ interactions during embryogenesis and disease.
Single-cell transcriptomics technology
The most established single-cell histological approach, single-cell transcriptomics technology, has recently begun to examine organogenesis with unprecedented resolution. Specifically, it projects the transcriptional profile of the paracrine signaling pathway into the progenitor gene lineage, inferring a roadmap of endoderm and mesoderm-induced interactions that orchestrate organogenesis [148]. Additionally, single-cell transcriptomics can model intra-organ crosstalk and chemotaxis-directed effects in the cardiac lineage, detecting patterning processes during human cardiac development [87, 149]. The full-cycle multi-omics map of cardiac development has been constructed by multi-omics technology [150]. A high-resolution spatial single-cell map of the developing human heart has been created by integrating single-cell sequencing with high-resolution in situ hybridization [151]. This map serves as a platform for drug screening and multiorgan systemic disease treatment by pooling expression communities across different organs to construct a comprehensive multi-organ profile of the cardiac-organ axis.
Single-cell transcriptomics allows for highly resolved, revealing the development and fate of lineages in organogenesis. Current genealogical recorders that combine reporter barcodes [152] with inducible CRISPR/Cas9 [153] can dynamically track clones within the initial hiPSCs pool. These methods map the dynamic states of multilineage organoids through detailed cellular and molecular roadmaps of cell fates [154]. Moreover, simulations of cellular movements and germ layer interactions in the formation of zebrafish protocorm embryos, achieved through single-cell transcriptome and in situ hybridization analyses, provide insights into head formation [155]. These approaches offer profound insights into lineage specification coordination and associated paracrine signaling, enhancing our understanding of the complex processes regulating inter-organ communications during embryogenesis.
Spatial transcriptomics technology
Cardiovascular diseases are the leading cause of death worldwide, with high morbidity and mortality rates. Emerging spatial transcriptomics techniques enable the elucidation of cellular spatial localization and the construction of dynamic 3D single-cell transcriptional profiles. This approach reveals the cellular spatial location and structure in the continuous progression from angina pectoris to myocardial infarction to heart failure, ultimately identifying new therapeutic targets [154]. In addition, the current spatiotemporal transcriptional profiles involve precise interactions between cell types in the three germ layers of gut development, which map all gut compartments and highlight coordinated events and crosstalk effects [156]. Notably, single-cell and spatial transcriptomic analysis allows for a comprehensive classification of neurons and neuronal circuits in the brain, enabling the reconstruction of multiple temporal and spatial dimensions. By integrating metabolomic profiles with genetic and epigenetic modifications, this approach offers new insights into the mechanisms underlying neurological disorders [157].
Spatial transcriptomics technology demonstrates the spatiotemporal mapping of human embryonic development and various multi-organ diseases, providing precise localization of gene expression and intercellular interactions. The capability is beneficial for presenting the key regulatory pathways of specific molecular interactions in multi-organ diseases [158]. Spatial transcriptomics provides new insights into disease genesis and elucidates changes in spatiotemporal effects among different organs at the multi-organ level.
Metabolomics technology
Organoids offer an ideal platform for drug screening, and exploring metabolomic regulation or metabolite transport mechanisms under both normal and pathological conditions. In particular, patient-derived organoids are invaluable for tailored personalized treatments, as they enable the identification of specific or optimal drugs for individual diseases or patients [159].
Mass spectrometry (MS) remains the cornerstone of metabolomics due to its unparalleled sensitivity, selectivity and capability for identifying metabolites. Among the available methods, liquid chromatography-mass spectrometry (LC-MS) is the most widely used for studying metabolomic regulatory mechanisms in complex biological systems. LC-MS facilitates the analysis of intricate mixtures such as organoids, allowing researchers to map metabolic pathways, discover biomarkers, and better understand disease progression [160]. Additionally, gas chromatography-mass spectrometry complements LC-MS by enabling targeted analysis of specific drugs and their metabolites.
Recent advancements have extended metabolomics to investigate interactions between the gut and brain axis interactions through complex organoid models [161]. In microbe-host interaction studies, changes in the microbial populations with related to metabolites such as brain neurotransmitters can be quantified using high-throughput LC-MS techniques, providing valuable insights into neurological diseases. Furthermore, the influence of the gut microbiome on cardiovascular health is increasingly evident. Short-chain fatty acids produced by gut microbes are known to regulate blood pressure through various receptors and pathways [162]. Studying the pathological responses of cardiac organoids to short-chain fatty acid stimulation offers a promising avenue for understanding the gut-heart axis and the mechanisms underlying CVDs.
Metabolomics in organoids also represents exciting possibilities for integrative systems biology. By integrating metabolomics with multi-omics technologies, including whole-genome sequencing, transcriptomics, ATAC-seq, and untargeted metabolomics, a multi-dimensional data platform can be developed. This platform enables a more comprehensive understanding of inter-organ communications in the human body and advances the potential of personalized medicine applications [163].
Proteomics technology
Proteomics analysis offers a powerful tool for uncovering intercellular signaling pathways and functional protein networks in health and disease. When applied to 3D organoid models derived from human cells, proteomics provides profound insights into the molecular mechanisms underlying clinical conditions [164].
Several primary approaches are currently used in proteomics. The most widely employed method is high-throughput analysis via MS, which relies on liquid chromatography-chromatography and signal capture based on fragmented peptide sequences and spectral counting [165]. This technique can identify thousands of proteins, allowing researchers to target key pathways effectively. Antigen-antibody array technology is suitable for medium- to low-weight proteomics analysis. It captures signals from fluorescence, chemiluminescence, or oligonucleotide-coupled tag labeling [166]. This method enables the study of protein-protein, lipid, small molecule, nucleic acid and antibody interactions. Finally, reversed-phase protein array technology facilitates large-scale protein sample analysis. It captures quantitative signals through colorimetric amplification or fluorescence detection, making it suitable for tracking proteins in signaling networks and studying post-translational modifications, including phosphorylation, methylation, and acetylation [167].
Organoid systems incorporating multiple cell populations further enrich proteomics research by closely mimicking the complexity of human tissues. Multidimensional proteomics analysis, combined with innovative predictive algorithms, will provide a more accurate representation of organism complexity in disease environments, holding great promise for advancing clinical drug development and therapy [168]. As organoid technologies evolve, the scalability and reproducibility of proteomics applications are expected to be improved. These advancements will provide unprecedented opportunities for drug development, biomarker discovery and the design of targeted therapies, ultimately translating into better clinical outcomes.
Conclusion and perspectives
This review illustrates the application of hiPSC-derived models for studying interactions between the heart and other organs at both 2D and 3D levels. We also introduce advanced tools for analyzing these interactions, focusing on the coordinated interactions in organ development. Critical methodologies include single-cell and spatial transcriptomics, which identify potential action targets and assess the reciprocal effects between the heart and other organs (Fig. 2).
Advances in heart-organ interaction studies using hiPSC-derived 2D and 3D models. With advancements in hiPSC-derived 2D cell co-culture systems, 3D humanoid organoids and microfluidic chip technologies, alongside emerging high-throughput sequencing technologies, it is now possible to investigate heart-organ communications in heart-involved multiple organ dysfunction syndromes in vitro. Created in BioRender. Ni, B. (2024) BioRender.com/a92e049
When employing 2D cell cultures or 3D organoid models, extensive measures are necessary to improve cell maturation and overall tissue complexity to mimic human physiology and pathology accurately. Researchers are continuously exploring novel organoid models and innovative practices that combine organoids with the immune system, as summarized in a recent review [169]. With the advancement of microfluidic devices, real-time imaging, and 3D printing, combining organoids with co-culture strategy will further deepen researchers’ understanding of organ development, disease onset, and progression. Newly developed organoid chip technologies show great potential for constructing higher fidelity organ models, and modern Organoids-on-a-chip systems hold great promise for elucidating the mechanisms of systemic diseases [170, 171].
Despite their potential, organ microarrays are still relatively new in CVD research, with limited studies on cardiac organoids. Many CVDs require modeling their complexity at the multi-organ level, especially heart-brain level interactions, which affect inter-organ crosstalk in an interrelated manner through neurotransmission [172]. Transplantation of cardiac organoids has been carried out, and the highlight of cardiac organoid transplantation is the effect of enhancing the maturation of cardiomyocytes to achieve a molecular, structural, and physiological phenotype similar to that of the adult heart [173]. Notably, researchers have demonstrated that cardiac organoid transplants can restore the contractile function of injured primate hearts [174]. Future challenges may include the integration of biosensors and the development of complex constructs for cardiac-engineered tissues. We anticipate that the ongoing development of hiPSC-based organoid models and advanced technologies will further enhance our understanding of CVDs.
Data availability
Data sharing is not applicable to this article as no new data were created or analysed in this study.
Abbreviations
- 2D:
-
Two-dimensional
- 3D:
-
Three-dimensional
- AI:
-
Artificial intelligence
- CCIs:
-
Cell-cell interactions
- CX43:
-
Connexin 43
- ECM:
-
Extracellular matrix
- ICAM-1:
-
Intercellular cell adhesion molecule-1
- LC-MS:
-
Liquid chromatography-mass spectrometry
- IL-1β:
-
Interleukin-1beta
- iPSC-CMs:
-
Induced pluripotent stem cell-derived cardiomyocytes
- MS:
-
Mass spectrometry
- ROS:
-
Reactive oxygen species
- SCN1A:
-
Sodium voltage-gated channel alpha subunit 1
- αSMA:
-
Alpha-smooth muscle actin
- TNF-α:
-
Tumor necrosis factor alpha
- VEGF:
-
Vascular endothelial growth factor
References
Doran S, Arif M, Lam S, Bayraktar A, Turkez H, Uhlen M, et al. Multi-omics approaches for revealing the complexity of cardiovascular disease. Brief Bioinform. 2021;22(5):bbab061.
Fujiu K, Shibata M, Nakayama Y, Ogata F, Matsumoto S, Noshita K, et al. A heart-brain-kidney network controls adaptation to cardiac stress through tissue macrophage activation. Nat Med. 2017;23(5):611–22.
Quinton LJ, Walkey AJ, Mizgerd JP. Integrative Physiology of Pneumonia. Physiol Rev. 2018;98(3):1417–64.
Duell PB, Welty FK, Miller M, Chait A, Hammond G, Ahmad Z, et al. Nonalcoholic Fatty Liver Disease and Cardiovascular Risk: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol. 2022;42(6):e168–85.
Witkowski M, Weeks TL, Hazen SL. Gut Microbiota and Cardiovascular Disease. Circ Res. 2020;127(4):553–70.
Bucciarelli V, Caterino AL, Bianco F, Caputi CG, Salerni S, Sciomer S, et al. Depression and cardiovascular disease: The deep blue sea of women’s heart. Trends Cardiovasc Med. 2020;30(3):170–6.
Stakos DA, Stamatelopoulos K, Bampatsias D, Sachse M, Zormpas E, Vlachogiannis NI, et al. The Alzheimer’s Disease Amyloid-Beta Hypothesis in Cardiovascular Aging and Disease: JACC Focus Seminar. J Am Coll Cardiol. 2020;75(8):952–67.
Miethe S, Karsonova A, Karaulov A, Renz H. Obesity and asthma. J Allergy Clin Immunol. 2020;146(4):685–93.
Lieder HR, Paket U, Skyschally A, Rink AD, Baars T, Neuhäuser M et al. Vago-splenic signal transduction of cardioprotection in humans. Eur Heart J. 2024:ehae250.
Yang Z, Zhang Y, Wang J, Yin J, Wang Z, Pei R. Cardiac organoid: multiple construction approaches and potential applications. J Mater Chem B. 2023;11(32):7567–81.
Majd H, Amin S, Ghazizadeh Z, Cesiulis A, Arroyo E, Lankford K, et al. Deriving Schwann cells from hPSCs enables disease modeling and drug discovery for diabetic peripheral neuropathy. Cell Stem Cell. 2023;30(5):632–e4710.
Van Lent J, Prior R, Pérez Siles G, Cutrupi AN, Kennerson ML, Vangansewinkel T, et al. Advances and challenges in modeling inherited peripheral neuropathies using iPSCs. Exp Mol Med. 2024;56(6):1348–64.
Kishimoto K, Iwasawa K, Sorel A, Ferran-Heredia C, Han L, Morimoto M, et al. Directed differentiation of human pluripotent stem cells into diverse organ-specific mesenchyme of the digestive and respiratory systems. Nat Protoc. 2022;17(11):2699–719.
Sharma A, Sances S, Workman MJ, Svendsen CN. Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell. 2020;26(3):309–29.
O’Brien LE. Tissue Homeostasis and Non-Homeostasis: From Cell Life Cycles to Organ States. Annu Rev Cell Dev Biol. 2022;38:395–418.
Kerkering J, Muinjonov B, Rosiewicz KS, Diecke S, Biese C, Schiweck J, et al. iPSC-derived reactive astrocytes from patients with multiple sclerosis protect cocultured neurons in inflammatory conditions. J Clin Invest. 2023;133(13):e164637.
Wang BX, Kit-Anan W, Terracciano CM. Many cells make life work—multicellularity in stem cell-based cardiac disease modelling. Int J Mol Sci. 2018;19(11):3361.
Maheshwari S, Vilema-Enríquez G, Wade-Martins R. Patient-derived iPSC models of Friedreich ataxia: a new frontier for understanding disease mechanisms and therapeutic application. Translational Neurodegeneration. 2023;12(1):45.
Habecker BA, Bers DM, Birren SJ, Chang R, Herring N, Kay MW et al. Molecular and cellular neurocardiology in heart disease. J Physiol. 2024.
Kowalski WJ, Garcia-Pak IH, Li W, Uosaki H, Tampakakis E, Zou J, et al. Sympathetic Neurons Regulate Cardiomyocyte Maturation in Culture. Front Cell Dev Biol. 2022;10:850645.
Wu HF, Saito-Diaz K, Huang CW, McAlpine JL, Seo DE, Magruder DS, et al. Parasympathetic neurons derived from human pluripotent stem cells model human diseases and development. Cell Stem Cell. 2024;31(5):734–e538.
Jian Y, Zhou X, Shan W, Chen C, Ge W, Cui J, et al. Crosstalk between macrophages and cardiac cells after myocardial infarction. Cell Commun Signal. 2023;21(1):109.
Sansonetti M, Al Soodi B, Thum T, Jung M. Macrophage-based therapeutic approaches for cardiovascular diseases. Basic Res Cardiol. 2024;119(1):1–33.
Wagner MJ, Khan M, Mohsin S. Healing the Broken Heart; The Immunomodulatory Effects of Stem Cell Therapy. Front Immunol. 2020;11:639.
Hitscherich PG, Xie LH, Del Re D, Lee EJ. The effects of macrophages on cardiomyocyte calcium-handling function using in vitro culture models. Physiol Rep. 2019;7(13):e14137.
Koc A, Akdeniz C, Cagavi E. Human macrophages directly modulate iPSC-derived cardiomyocytes at healthy state and congenital arrhythmia model in vitro. Pflugers Arch. 2022;474(12):1295–310.
Liu C, Liu Y, Chen H, Yang X, Lu C, Wang L, et al. Myocardial injury: where inflammation and autophagy meet. Burns Trauma. 2023;11:tkac062.
Jia D, Chen S, Bai P, Luo C, Liu J, Sun A, et al. Cardiac Resident Macrophage-Derived Legumain Improves Cardiac Repair by Promoting Clearance and Degradation of Apoptotic Cardiomyocytes After Myocardial Infarction. Circulation. 2022;145(20):1542–56.
Talukdar D, Haldar AK, Kumar S, Dastidar R, Basu A, Roy A, et al. Leukocyte infiltration and cross-talk with cardiomyocytes exploit intracellular stress pathways in dilated cardiomyopathy of idiopathic origin. Mol Biol Rep. 2024;51(1):1090.
Rauchenwald T, Handle F, Connolly CE, Degen A, Seifarth C, Hermann M, et al. Preadipocytes in human granulation tissue: role in wound healing and response to macrophage polarization. Inflamm Regen. 2023;43(1):53.
Watanabe H, Kanemaru K, Hagikura K, Matsumoto T, Ayusawa M, Morioka I. Soluble factors released by dedifferentiated fat cells reduce the functional activity of iPS cell-derived cardiomyocytes. Cell Biol Int. 2021;45(2):295–304.
Fukuma N, Tokiwa H, Numata G, Ueda K, Liu PY, Tajima M, et al. Endothelial oestrogen-myocardial cyclic guanosine monophosphate axis critically determines angiogenesis and cardiac performance during pressure overload. Cardiovasc Res. 2024;120(15):1884–97.
Suarez-Martinez E, Suazo-Sanchez I, Celis-Romero M, Carnero A. 3D and organoid culture in research: Physiology, hereditary genetic diseases and cancer. Cell Bioscience. 2022;12(1):1–19.
Sun L, Bian F, Xu D, Luo Y, Wang Y, Zhao Y. Tailoring biomaterials for biomimetic organs-on-chips. Mater Horiz. 2023;10(11):4724–45.
Park SE, Georgescu A, Huh D. Organoids-on-a-chip. Science. 2019;364(6444):960–5.
Xie R, Pal V, Yu Y, Lu X, Gao M, Liang S, et al. A comprehensive review on 3D tissue models: Biofabrication technologies and preclinical applications. Biomaterials. 2024;304:122408.
Ouchi R, Togo S, Kimura M, Shinozawa T, Koido M, Koike H, et al. Modeling Steatohepatitis in Humans with Pluripotent Stem Cell-Derived Organoids. Cell Metab. 2019;30(2):374–84.e6.
Du X, Jia H, Chang Y, Zhao Y, Song J. Progress of organoid platform in cardiovascular research. Bioact Mater. 2024;40:88–103.
Zhi Y, Zhu Y, Wang J, Zhao J, Zhao Y. Cortical Organoid-on-a-Chip with Physiological Hypoxia for Investigating Tanshinone IIA-Induced Neural Differentiation. Research (Wash D C). 2023;6:0273.
Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discovery. 2021;20(5):345–61.
Vunjak-Novakovic G, Ronaldson-Bouchard K, Radisic M. Organs-on-a-chip models for biological research. Cell. 2021;184(18):4597–611.
Lewis-Israeli YR, Wasserman AH, Gabalski MA, Volmert BD, Ming Y, Ball KA, et al. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat Commun. 2021;12(1):5142.
Ng WH, Varghese B, Jia H, Ren X. Alliance of Heart and Endoderm: Multilineage Organoids to Model Co-development. Circ Res. 2023;132(4):511–8.
Branco MA, Dias TP, Cabral JMS, Pinto-do-Ó P, Diogo MM. Human multilineage pro-epicardium/foregut organoids support the development of an epicardium/myocardium organoid. Nat Commun. 2022;13(1):6981.
Gu M, Zorn AM. Follow your heart and trust your gut: Co-development of heart and gut tissue in organoids. Cell Stem Cell. 2021;28(12):2037–8.
Rossi G, Broguiere N, Miyamoto M, Boni A, Guiet R, Girgin M, et al. Capturing Cardiogenesis in Gastruloids. Cell Stem Cell. 2021;28(2):230–e406.
Silva AC, Matthys OB, Joy DA, Kauss MA, Natarajan V, Lai MH, et al. Co-emergence of cardiac and gut tissues promotes cardiomyocyte maturation within human iPSC-derived organoids. Cell Stem Cell. 2021;28(12):2137–e526.
Wang Y, Qin J. Advances in human organoids-on-chips in biomedical research. Life Med. 2023;2(1):lnad007.
Tao T, Deng P, Wang Y, Zhang X, Guo Y, Chen W, et al. Microengineered Multi-Organoid System from hiPSCs to Recapitulate Human Liver-Islet Axis in Normal and Type 2 Diabetes. Adv Sci (Weinh). 2022;9(5):e2103495.
Faustino Martins JM, Fischer C, Urzi A, Vidal R, Kunz S, Ruffault PL, et al. Self-Organizing 3D Human Trunk Neuromuscular Organoids. Cell Stem Cell. 2020;26(2):172–e866.
Kelley KW, Pașca SP. Human brain organogenesis: Toward a cellular understanding of development and disease. Cell. 2022;185(1):42–61.
Vogt N, Assembloids. Nat Methods. 2021;18(1):27.
Kanton S, Paşca SP. Human assembloids. Development. 2022;149(20):dev201120.
Li J, Wiesinger A, Fokkert L, Bakker P, de Vries DK, Tijsen AJ, et al. Modeling the atrioventricular conduction axis using human pluripotent stem cell-derived cardiac assembloids. Cell Stem Cell. 2024;31(11):1667–e846.
Zeltner N, Wu HF, Saito-Diaz K, Sun X, Song M, Saini T et al. A modular platform to generate functional sympathetic neuron-innervated heart assembloids. Res Sq. 2024;rs(3):rs-3894397.
Rockel AF, Wagner N, Spenger P, Ergün S, Wörsdörfer P. Neuro-mesodermal assembloids recapitulate aspects of peripheral nervous system development in vitro. Stem Cell Rep. 2023;18(5):1155–65.
Yang J, Lei W, Xiao Y, Tan S, Yang J, Lin Y, et al. Generation of human vascularized and chambered cardiac organoids for cardiac disease modelling and drug evaluation. Cell Prolif. 2024;57(8):e13631.
Ng WH, Johnston EK, Tan JJ, Bliley JM, Feinberg AW, Stolz DB et al. Recapitulating human cardio-pulmonary co-development using simultaneous multilineage differentiation of pluripotent stem cells. Elife. 2022; 11.
Boylin K, Aquino GV, Purdon M, Abedi K, Kasendra M, Barrile R. Basic models to advanced systems: harnessing the power of organoids-based microphysiological models of the human brain. Biofabrication. 2024;16(3):032007.
Andersen J, Revah O, Miura Y, Thom N, Amin ND, Kelley KW, et al. Generation of functional human 3D cortico-motor assembloids. Cell. 2020;183(7):1913–29.e26.
Wolf KJ, Weiss JD, Uzel SGM, Skylar-Scott MA, Lewis JA. Biomanufacturing human tissues via organ building blocks. Cell Stem Cell. 2022;29(5):667–77.
Leng Y, Li X, Zheng F, Liu H, Wang C, Wang X, et al. Advances in In Vitro Models of Neuromuscular Junction: Focusing on Organ-on-a-Chip, Organoids, and Biohybrid Robotics. Adv Mater. 2023;35(41):e2211059.
Cakir B, Xiang Y, Tanaka Y, Kural MH, Parent M, Kang YJ, et al. Engineering of human brain organoids with a functional vascular-like system. Nat Methods. 2019;16(11):1169–75.
Mabry SA, Pavon N. Exploring the prospects, advancements, and challenges of in vitro modeling of the heart-brain axis. Front Cell Neurosci. 2024;18:1386355.
Onesto MM, Kim JI, Pasca SP. Assembloid models of cell-cell interaction to study tissue and disease biology. Cell Stem Cell. 2024;31(11):1563–73.
Kim H, Kamm RD, Vunjak-Novakovic G, Wu JC. Progress in multicellular human cardiac organoids for clinical applications. Cell Stem Cell. 2022;29(4):503–14.
Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G, Maruccio G. Organs-on-chips technologies - A guide from disease models to opportunities for drug development. Biosens Bioelectron. 2023;231:115271.
Palasantzas V, Tamargo-Rubio I, Le K, Slager J, Wijmenga C, Jonkers IH, et al. iPSC-derived organ-on-a-chip models for personalized human genetics and pharmacogenomics studies. Trends Genet. 2023;39(4):268–84.
Huang Y, Liu T, Huang Q, Wang Y. From Organ-on-a-Chip to Human-on-a-Chip: A Review of Research Progress and Latest Applications. ACS Sens. 2024;9(7):3466–88.
Saorin G, Caligiuri I, Rizzolio F. Microfluidic organoids-on-a-chip: The future of human models. Semin Cell Dev Biol. 2023;144:41–54.
Man Y, Liu Y, Chen Q, Zhang Z, Li M, Xu L, et al. Organoids-On-a-Chip for Personalized Precision Medicine. Adv Healthc Mater. 2024;13(30):e2401843.
Wu L, Ai Y, Xie R, Xiong J, Wang Y, Liang Q. Organoids/organs-on-a-chip: new frontiers of intestinal pathophysiological models. Lab Chip. 2023;23(5):1192–212.
Deng AF, Wang FX, Wang SC, Zhang YZ, Bai L, Su JC. Bone-organ axes: bidirectional crosstalk. Mil Med Res. 2024;11(1):37.
Vandernoot I, Haerlingen B, Gillotay P, Trubiroha A, Janssens V, Opitz R, et al. Enhanced Canonical Wnt Signaling During Early Zebrafish Development Perturbs the Interaction of Cardiac Mesoderm and Pharyngeal Endoderm and Causes Thyroid Specification Defects. Thyroid. 2021;31(3):420–38.
Han L, Chaturvedi P, Kishimoto K, Koike H, Nasr T, Iwasawa K, et al. Single cell transcriptomics identifies a signaling network coordinating endoderm and mesoderm diversification during foregut organogenesis. Nat Commun. 2020;11(1):4158.
Koike H, Iwasawa K, Ouchi R, Maezawa M, Giesbrecht K, Saiki N, et al. Modelling human hepato-biliary-pancreatic organogenesis from the foregut-midgut boundary. Nature. 2019;574(7776):112–6.
Shewale B, Dubois N. Of form and function: Early cardiac morphogenesis across classical and emerging model systems. Semin Cell Dev Biol. 2021;118:107–18.
Branco MA, Dias TP, Cabral JMS, Pinto-do OP, Diogo MM. Human multilineage pro-epicardium/foregut organoids support the development of an epicardium/myocardium organoid. Nat Commun. 2022;13(1):6981.
Drakhlis L, Biswanath S, Farr CM, Lupanow V, Teske J, Ritzenhoff K, et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat Biotechnol. 2021;39(6):737–46.
Liu D, Song AT, Qi X, van Vliet PP, Xiao J, Xiong F, et al. Cohesin-protein Shugoshin-1 controls cardiac automaticity via HCN4 pacemaker channel. Nat Commun. 2021;12(1):2551.
Qi H, Liu H, Pullamsetti SS, Günther S, Kuenne C, Atzberger A, et al. Epigenetic regulation by Suv4-20h1 in cardiopulmonary progenitor cells is required to prevent pulmonary hypertension and chronic obstructive pulmonary disease. Circulation. 2021;144(13):1042–58.
Steimle JD, Rankin SA, Slagle CE, Bekeny J, Rydeen AB, Chan SS, et al. Evolutionarily conserved Tbx5-Wnt2/2b pathway orchestrates cardiopulmonary development. Proc Natl Acad Sci U S A. 2018;115(45):E10615–24.
Rankin SA, Steimle JD, Yang XH, Rydeen AB, Agarwal K, Chaturvedi P, et al. Tbx5 drives Aldh1a2 expression to regulate a RA-Hedgehog-Wnt gene regulatory network coordinating cardiopulmonary development. Elife. 2021;10:e69288.
Ng WH, Johnston EK, Tan JJ, Bliley JM, Feinberg AW, Stolz DB, et al. Recapitulating human cardio-pulmonary co-development using simultaneous multilineage differentiation of pluripotent stem cells. Elife. 2022;11:e67872.
Xia L-x, Xiao Y-y, Jiang W-j, Yang X-y, Tao H, Mandukhail SR et al. Exosomes derived from induced cardiopulmonary progenitor cells alleviate acute lung injury in mice. Acta Pharmacol Sin. 2024:1–16.
Chatterjee D, Ing RJ, Gien J. Update on Congenital Diaphragmatic Hernia. Anesth Analg. 2020;131(3):808–21.
Meier AB, Zawada D, De Angelis MT, Martens LD, Santamaria G, Zengerle S, et al. Epicardioid single-cell genomics uncovers principles of human epicardium biology in heart development and disease. Nat Biotechnol. 2023;41(12):1787–800.
Kidokoro H, Yonei-Tamura S, Tamura K, Schoenwolf GC, Saijoh Y. The heart tube forms and elongates through dynamic cell rearrangement coordinated with foregut extension. Development. 2018;145(7):dev152488.
Zhang J, Zhu P, Li S, Gao Y, Xing Y. From heart failure and kidney dysfunction to cardiorenal syndrome: TMAO may be a bridge. Front Pharmacol. 2023;14:1291922.
Young JB, Eknoyan G. Cardiorenal Syndrome: An Evolutionary Appraisal. Circ Heart Fail. 2024;17(6):e011510.
Xu Z, Yang J, Xin X, Liu C, Li L, Mei X, et al. Merits and challenges of iPSC-derived organoids for clinical applications. Front Cell Dev Biol. 2023;11:1188905.
Gabbin B, Meraviglia V, Angenent ML, Ward-van Oostwaard D, Sol W, Mummery CL, et al. Heart and kidney organoids maintain organ-specific function in a microfluidic system. Mater Today Bio. 2023;23:100818.
Song SS, Park HJ, Kim YK, Kang SW. Revolutionizing biomedical research: The imperative need for heart-kidney-connected organoids. APL Bioeng. 2024;8(1):010902.
Wijarnpreecha K, Aby ES, Ahmed A, Kim D. Evaluation and management of extrahepatic manifestations of nonalcoholic fatty liver disease. Clin Mol Hepatol. 2021;27(2):221–35.
Yin F, Zhang X, Wang L, Wang Y, Zhu Y, Li Z, et al. HiPSC-derived multi-organoids-on-chip system for safety assessment of antidepressant drugs. Lab Chip. 2021;21(3):571–81.
Oleaga C, Riu A, Rothemund S, Lavado A, McAleer CW, Long CJ, et al. Investigation of the effect of hepatic metabolism on off-target cardiotoxicity in a multi-organ human-on-a-chip system. Biomaterials. 2018;182:176–90.
Ferrari E, Rasponi M. Liver-Heart on chip models for drug safety. APL Bioeng. 2021;5(3):031505.
Chakraborty P, Farhat K, Po SS, Armoundas AA, Stavrakis S. Autonomic Nervous System and Cardiac Metabolism: Links Between Autonomic and Metabolic Remodeling in Atrial Fibrillation. JACC Clin Electrophysiol. 2023;9(7 Pt 2):1196–206.
Bernardin AA, Colombani S, Rousselot A, Andry V, Goumon Y, Delanoë-Ayari H, et al. Impact of neurons on patient-derived cardiomyocytes using organ-On-A-Chip and iPSC biotechnologies. Cells. 2022;11(23):3764.
Huynh K. Heartbeat-induced pressure pulsations in cerebral arteries modulate neuronal activity. Nat Rev Cardiol. 2024;21(4):218.
Jammal Salameh L, Bitzenhofer SH, Hanganu-Opatz IL, Dutschmann M, Egger V. Blood pressure pulsations modulate central neuronal activity via mechanosensitive ion channels. Science. 2024;383(6682):eadk8511.
Manolis AA, Manolis TA, Apostolopoulos EJ, Apostolaki NE, Melita H, Manolis AS. The role of the autonomic nervous system in cardiac arrhythmias: The neuro-cardiac axis, more foe than friend? Trends Cardiovasc Med. 2021;31(5):290–302.
Herring N, Kalla M, Paterson DJ. The autonomic nervous system and cardiac arrhythmias: current concepts and emerging therapies. Nat Rev Cardiol. 2019;16(12):707–26.
Butler D, Reyes DR. Heart-on-a-chip systems: disease modeling and drug screening applications. Lab Chip. 2024;24(5):1494–528.
Skardal A, Murphy SV, Devarasetty M, Mead I, Kang HW, Seol YJ, et al. Multi-tissue interactions in an integrated three-tissue organ-on-a-chip platform. Sci Rep. 2017;7(1):8837.
Rajan SAP, Aleman J, Wan M, Pourhabibi Zarandi N, Nzou G, Murphy S, et al. Probing prodrug metabolism and reciprocal toxicity with an integrated and humanized multi-tissue organ-on-a-chip platform. Acta Biomater. 2020;106:124–35.
Ronaldson-Bouchard K, Teles D, Yeager K, Tavakol DN, Zhao Y, Chramiec A, et al. A multi-organ chip with matured tissue niches linked by vascular flow. Nat Biomed Eng. 2022;6(4):351–71.
Vogt N. Modeling multi-organ systems on a chip. Nat Methods. 2022;19(6):641.
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L, et al. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci. 2024;25(17):9186.
Li Y, Li C, Liu Q, Wang L, Bao AX, Jung JP, et al. Loss of Acta2 in cardiac fibroblasts does not prevent the myofibroblast differentiation or affect the cardiac repair after myocardial infarction. J Mol Cell Cardiol. 2022;171:117–32.
Feyen DAM, McKeithan WL, Bruyneel AAN, Spiering S, Hörmann L, Ulmer B, et al. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep. 2020;32(3):107925.
Garg P, Garg V, Shrestha R, Sanguinetti MC, Kamp TJ, Wu JC. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists. Circ Res. 2018;123(2):224–43.
Karbassi E, Fenix A, Marchiano S, Muraoka N, Nakamura K, Yang X, et al. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat Rev Cardiol. 2020;17(6):341–59.
Zhang H, Pei L, Ouyang Z, Wang H, Chen X, Jiang K, et al. AP-1 activation mediates post-natal cardiomyocyte maturation. Cardiovasc Res. 2023;119(2):536–50.
Wickramasinghe NM, Sachs D, Shewale B, Gonzalez DM, Dhanan-Krishnan P, Torre D, et al. PPARdelta activation induces metabolic and contractile maturation of human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell. 2022;29(4):559–e767.
Pabel S, Reetz F, Dybkova N, Shomroni O, Salinas G, Mustroph J, et al. Long-term effects of empagliflozin on excitation-contraction-coupling in human induced pluripotent stem cell cardiomyocytes. J Mol Med (Berl). 2020;98(12):1689–700.
Ronaldson-Bouchard K, Ma SP, Yeager K, Chen T, Song L, Sirabella D, et al. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature. 2018;556(7700):239–43.
Fetterman KA, Blancard M, Lyra-Leite DM, Vanoye CG, Fonoudi H, Jouni M, et al. Independent compartmentalization of functional, metabolic, and transcriptional maturation of hiPSC-derived cardiomyocytes. Cell Rep. 2024;43(5):114160.
Mozneb M, Jenkins A, Sances S, Pohlman S, Workman MJ, West D, et al. Multi-lineage heart-chip models drug cardiotoxicity and enhances maturation of human stem cell-derived cardiovascular cells. Lab Chip. 2024;24(4):869–81.
Lu K, Seidel T, Cao-Ehlker X, Dorn T, Batcha AMN, Schneider CM, et al. Progressive stretch enhances growth and maturation of 3D stem-cell-derived myocardium. Theranostics. 2021;11(13):6138–53.
Ruan JL, Tulloch NL, Razumova MV, Saiget M, Muskheli V, Pabon L, et al. Mechanical Stress Conditioning and Electrical Stimulation Promote Contractility and Force Maturation of Induced Pluripotent Stem Cell-Derived Human Cardiac Tissue. Circulation. 2016;134(20):1557–67.
Giacomelli E, Meraviglia V, Campostrini G, Cochrane A, Cao X, van Helden RWJ, et al. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease. Cell Stem Cell. 2020;26(6):862–e7911.
Yoshida S, Miyagawa S, Fukushima S, Kawamura T, Kashiyama N, Ohashi F, et al. Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes by Soluble Factors from Human Mesenchymal Stem Cells. Mol Ther. 2018;26(11):2681–95.
Yang H, Yang Y, Kiskin FN, Shen M, Zhang JZ. Recent advances in regulating the proliferation or maturation of human-induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther. 2023;14(1):228.
Shadrin IY, Allen BW, Qian Y, Jackman CP, Carlson AL, Juhas ME, et al. Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat Commun. 2017;8(1):1825.
Lin Z, Garbern JC, Liu R, Li Q, Mancheño Juncosa E, Elwell HLT, et al. Tissue-embedded stretchable nanoelectronics reveal endothelial cell-mediated electrical maturation of human 3D cardiac microtissues. Sci Adv. 2023;9(10):eade8513.
Magalhães S, Almeida I, Pereira CD, Rebelo S, Goodfellow BJ, Nunes A. The Long-Term Culture of Human Fibroblasts Reveals a Spectroscopic Signature of Senescence. Int J Mol Sci. 2022; 23(10).
Song M, Choi DB, Im JS, Song YN, Kim JH, Lee H, et al. Modeling acute myocardial infarction and cardiac fibrosis using human induced pluripotent stem cell-derived multi-cellular heart organoids. Cell Death Dis. 2024;15(5):308.
Richards DJ, Li Y, Kerr CM, Yao J, Beeson GC, Coyle RC, et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng. 2020;4(4):446–62.
Wu Y, Wang Y, Xiao M, Zhang G, Zhang F, Tang M et al. 3D-Printed Myocardium-Specific Structure Enhances Maturation and Therapeutic Efficacy of Engineered Heart Tissue in Myocardial Infarction. Adv Sci (Weinh). 2025:e2409871.
Ewoldt JK, DePalma SJ, Jewett ME, Karakan M, Lin YM, Mir Hashemian P, et al. Induced pluripotent stem cell-derived cardiomyocyte in vitro models: benchmarking progress and ongoing challenges. Nat Methods. 2025;22(1):24–40.
Lee SG, Kim YJ, Son MY, Oh MS, Kim J, Ryu B, et al. Generation of human iPSCs derived heart organoids structurally and functionally similar to heart. Biomaterials. 2022;290:121860.
Dababneh S, Hamledari H, Maaref Y, Jayousi F, Hosseini DB, Khan A, et al. Advances in Hypertrophic Cardiomyopathy Disease Modelling Using hiPSC-Derived Cardiomyocytes. Can J Cardiol. 2024;40(5):766–76.
Johnson RD, Lei M, McVey JH, Camelliti P. Human myofibroblasts increase the arrhythmogenic potential of human induced pluripotent stem cell-derived cardiomyocytes. Cell Mol Life Sci. 2023;80(9):276.
van den Dolder FW, Dinani R, Warnaar VA, Vučković S, Passadouro AS, Nassar AA, et al. Experimental Models of Hypertrophic Cardiomyopathy: A Systematic Review. JACC: Basic to Translational Science; 2025.
Gandoy-Fieiras N, Gonzalez-Juanatey JR, Eiras S. Myocardium Metabolism in Physiological and Pathophysiological States: Implications of Epicardial Adipose Tissue and Potential Therapeutic Targets. Int J Mol Sci 2020; 21(7).
Ye L, Yu Y, Zhao ZA, Zhao D, Ni X, Wang Y, et al. Patient-specific iPSC-derived cardiomyocytes reveal abnormal regulation of FGF16 in a familial atrial septal defect. Cardiovasc Res. 2022;118(3):859–71.
Feng W, Schriever H, Jiang S, Bais A, Wu H, Kostka D, et al. Computational profiling of hiPSC-derived heart organoids reveals chamber defects associated with NKX2-5 deficiency. Commun Biology. 2022;5(1):399.
Chan CH, Lam YY, Wong N, Geng L, Zhang J, Ahola V, et al. Abnormal developmental trajectory and vulnerability to cardiac arrhythmias in tetralogy of Fallot with DiGeorge syndrome. Commun Biol. 2023;6(1):969.
Sallam K, Li Y, Sager PT, Houser SR, Wu JC. Finding the rhythm of sudden cardiac death: new opportunities using induced pluripotent stem cell-derived cardiomyocytes. Circ Res. 2015;116(12):1989–2004.
Fernández-Morales JC, Xia Y, Renzo TJ, Zhang XH, Morad M. Mutation in RyR2-FKBP Binding site alters Ca(2+) signaling modestly but increases arrhythmogenesis in human stem cells derived cardiomyocytes. Cell Calcium. 2022;101:102500.
Phang RJ, Ritchie RH, Hausenloy DJ, Lees JG, Lim SY. Cellular interplay between cardiomyocytes and non-myocytes in diabetic cardiomyopathy. Cardiovasc Res. 2023;119(3):668–90.
Kwok M, Lee C, Li HS, Deng R, Tsoi C, Ding Q, et al. Remdesivir induces persistent mitochondrial and structural damage in human induced pluripotent stem cell-derived cardiomyocytes. Cardiovasc Res. 2022;118(12):2652–64.
Fonoudi H, Jouni M, Cejas RB, Magdy T, Blancard M, Ge N, et al. Functional Validation of Doxorubicin-Induced Cardiotoxicity-Related Genes. JACC CardioOncol. 2024;6(1):38–50.
Baysoy A, Bai Z, Satija R, Fan R. The technological landscape and applications of single-cell multi-omics. Nat Rev Mol Cell Biol. 2023;24(10):695–713.
Moffitt JR, Lundberg E, Heyn H. The emerging landscape of spatial profiling technologies. Nat Rev Genet. 2022;23(12):741–59.
Vandereyken K, Sifrim A, Thienpont B, Voet T. Methods and applications for single-cell and spatial multi-omics. Nat Rev Genet. 2023;24(8):494–515.
Wang R, Yang X, Chen J, Zhang L, Griffiths JA, Cui G, et al. Time space and single-cell resolved tissue lineage trajectories and laterality of body plan at gastrulation. Nat Commun. 2023;14(1):5675.
Kelly RG. The heart field transcriptional landscape at single-cell resolution. Dev Cell. 2023;58(4):257–66.
Gu Y, Zhou Y, Ju S, Liu X, Zhang Z, Guo J, et al. Multi-omics profiling visualizes dynamics of cardiac development and functions. Cell Rep. 2022;41(13):111891.
Farah EN, Hu RK, Kern C, Zhang Q, Lu TY, Ma Q, et al. Spatially organized cellular communities form the developing human heart. Nature. 2024;627(8005):854–64.
Jindal K, Adil MT, Yamaguchi N, Yang X, Wang HC, Kamimoto K, et al. Single-cell lineage capture across genomic modalities with CellTag-multi reveals fate-specific gene regulatory changes. Nat Biotechnol. 2024;42(6):946–59.
He Z, Maynard A, Jain A, Gerber T, Petri R, Lin HC, et al. Lineage recording in human cerebral organoids. Nat Methods. 2022;19(1):90–9.
Han S, Xu Q, Du Y, Tang C, Cui H, Xia X, et al. Single-cell spatial transcriptomics in cardiovascular development, disease, and medicine. Genes Dis. 2024;11(6):101163.
Cheng T, Xing YY, Liu C, Li YF, Huang Y, Liu X, et al. Nodal coordinates the anterior-posterior patterning of germ layers and induces head formation in zebrafish explants. Cell Rep. 2023;42(4):112351.
Fawkner-Corbett D, Antanaviciute A, Parikh K, Jagielowicz M, Gerós AS, Gupta T, et al. Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell. 2021;184(3):810–e2623.
Wang W, Liu X, Wang DC. Single-cell and spatial alterations of neural cells and circuits in clinical and translational medicine. Clin Transl Med. 2024;14(6):e1696.
Zhang L, Chen D, Song D, Liu X, Zhang Y, Xu X, et al. Clinical and translational values of spatial transcriptomics. Signal Transduct Target Therapy. 2022;7(1):111.
Rauth S, Karmakar S, Batra SK, Ponnusamy MP. Recent advances in organoid development and applications in disease modeling. Biochim Biophys Acta Rev Cancer. 2021;1875(2):188527.
Khamis MM, Adamko DJ, El-Aneed A. Mass spectrometric based approaches in urine metabolomics and biomarker discovery. Mass Spectrom Rev. 2017;36(2):115–34.
Horvath TD, Haidacher SJ, Engevik MA, Luck B, Ruan W, Ihekweazu F, et al. Interrogation of the mammalian gut-brain axis using LC-MS/MS-based targeted metabolomics with in vitro bacterial and organoid cultures and in vivo gnotobiotic mouse models. Nat Protoc. 2023;18(2):490–529.
Xu J, Moore BN, Pluznick JL. Short-Chain Fatty Acid Receptors and Blood Pressure Regulation: Council on Hypertension Mid-Career Award for Research Excellence 2021. Hypertension. 2022;79(10):2127–37.
Zhu Y, Tang S, Yuan Q, Fu J, He J, Liu Z, et al. Integrated characterization of hepatobiliary tumor organoids provides a potential landscape of pharmacogenomic interactions. Cell Rep Med. 2024;5(2):101375.
Cristobal A, van den Toorn HWP, van de Wetering M, Clevers H, Heck AJR, Mohammed S. Personalized Proteome Profiles of Healthy and Tumor Human Colon Organoids Reveal Both Individual Diversity and Basic Features of Colorectal Cancer. Cell Rep. 2017;18(1):263–74.
Macklin A, Khan S, Kislinger T. Recent advances in mass spectrometry based clinical proteomics: applications to cancer research. Clin Proteom. 2020;17:17.
Reslova N, Michna V, Kasny M, Mikel P, Kralik P. xMAP Technology: Applications in Detection of Pathogens. Front Microbiol. 2017;8:55.
Torresano L, Santacatterina F, Domínguez-Zorita S, Nuevo-Tapioles C, Núñez-Salgado A, Esparza-Moltó PB, et al. Analysis of the metabolic proteome of lung adenocarcinomas by reverse-phase protein arrays (RPPA) emphasizes mitochondria as targets for therapy. Oncogenesis. 2022;11(1):24.
Huang P, Gao W, Fu C, Wang M, Li Y, Chu B et al. Clinical functional proteomics of intercellular signalling in pancreatic cancer. Nature. 2024.
Xu H, Jiao D, Liu A, Wu K. Tumor organoids: applications in cancer modeling and potentials in precision medicine. J Hematol Oncol. 2022;15(1):58.
Tang H, Abouleila Y, Si L, Ortega-Prieto AM, Mummery CL, Ingber DE, et al. Human Organs-on-Chips for Virology. Trends Microbiol. 2020;28(11):934–46.
Wang Y, Wang P, Qin J. Human Organoids and Organs-on-Chips for Addressing COVID-19 Challenges. Adv Sci (Weinh). 2022;9(10):e2105187.
Rossi A, Mikail N, Bengs S, Haider A, Treyer V, Buechel RR, et al. Heart-brain interactions in cardiac and brain diseases: why sex matters. Eur Heart J. 2022;43(39):3971–80.
Varzideh F, Pahlavan S, Ansari H, Halvaei M, Kostin S, Feiz MS, et al. Human cardiomyocytes undergo enhanced maturation in embryonic stem cell-derived organoid transplants. Biomaterials. 2019;192:537–50.
Kobayashi H, Tohyama S, Ichimura H, Ohashi N, Chino S, Soma Y, et al. Regeneration of Nonhuman Primate Hearts With Human Induced Pluripotent Stem Cell-Derived Cardiac Spheroids. Circulation. 2024;150(8):611–21.
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L et al. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci 2024; 25(17).
Acknowledgements
The authors declare that they have not use AI-generated work in this manuscript.
Funding
This work was supported by National Key R&D Program of China (2022YFA1104300, 2021YFA1101902), National Natural Science Foundation of China (82241202, 82170364, 81970223, 82200339), Natural Science Foundation of Jiangsu Province (BK20240001), the Space Medical Experiment Project of China Manned Space Program (HYZHXM01018), Jiangsu Cardiovascular Medicine Innovation Center (CXZX202210), National Center for International Research (2017B01012), and A Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions.
Author information
Authors and Affiliations
Contributions
B.N. and L.Y. reviewed the literature and drafted the manuscript; B.N. and Y. Z. designed the figures; S.H. and W.L. provided supervision and revised the manuscript. All authors approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Ni, B., Ye, L., Zhang, Y. et al. Advances in humanoid organoid-based research on inter-organ communications during cardiac organogenesis and cardiovascular diseases. J Transl Med 23, 380 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06381-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06381-x