Skip to content

This report is written by MaltSci based on the latest literature and research findings


How does cell-cell communication coordinate tissue function?

Abstract

Cell-cell communication (CCC) is a fundamental process that underpins the coordination of tissue function in multicellular organisms. This review systematically explores the mechanisms of CCC, emphasizing their roles in tissue development, homeostasis, and cellular response coordination. Key mechanisms include direct cell-cell interactions via gap junctions, paracrine signaling, and the release of extracellular vesicles, which facilitate localized and long-range communication. The review highlights the critical functions of CCC in tissue morphogenesis, repair mechanisms, and the implications of dysregulated communication in diseases such as cancer, inflammatory conditions, and neurodegenerative disorders. Notably, recent advancements in imaging techniques and molecular biology have unveiled the dynamic nature of cell interactions and the intricate signaling pathways that govern them. The identification of novel signaling molecules and bioelectrical controls in tissue dynamics further enriches our understanding of CCC. By synthesizing current research findings, this review aims to provide comprehensive insights into how cell-cell interactions influence tissue dynamics and to highlight potential therapeutic strategies targeting these pathways for disease intervention. Understanding the complexities of CCC not only enhances our knowledge of fundamental biological processes but also paves the way for innovative approaches in precision medicine and regenerative therapies.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Mechanisms of Cell-Cell Communication
    • 2.1 Direct Cell-Cell Interactions
    • 2.2 Paracrine Signaling
    • 2.3 Extracellular Vesicles and Signaling Molecules
  • 3 Role of Cell-Cell Communication in Tissue Function
    • 3.1 Tissue Development and Morphogenesis
    • 3.2 Homeostasis and Repair Mechanisms
    • 3.3 Coordination of Cellular Responses
  • 4 Dysregulation of Cell-Cell Communication in Disease
    • 4.1 Cancer
    • 4.2 Inflammatory Diseases
    • 4.3 Neurodegenerative Disorders
  • 5 Therapeutic Implications
    • 5.1 Targeting Communication Pathways in Cancer Therapy
    • 5.2 Modulating Cell-Cell Interactions for Tissue Regeneration
    • 5.3 Innovations in Drug Delivery via Extracellular Vesicles
  • 6 Future Directions in Research
    • 6.1 Advances in Imaging Techniques
    • 6.2 Exploring Novel Signaling Molecules
    • 6.3 Integrative Approaches to Study Tissue Dynamics
  • 7 Summary

1 Introduction

Cell-cell communication (CCC) is a fundamental process that underpins the coordination of tissue function in multicellular organisms. This intricate network of signaling pathways and molecular interactions enables cells to communicate and respond to their microenvironment, thereby maintaining homeostasis and facilitating tissue development, repair, and function. The ability of cells to effectively coordinate their activities is essential not only for normal physiological processes but also for the organism's overall health and well-being. Disruptions in these communication pathways can lead to a variety of diseases, underscoring the importance of understanding CCC in both health and disease contexts[1].

The significance of CCC extends beyond mere communication; it encompasses a variety of mechanisms, including direct interactions through gap junctions, paracrine signaling, and the release of extracellular vesicles. These mechanisms are vital for processes such as tissue morphogenesis, homeostasis, and the coordinated response of cells to external stimuli. For instance, recent studies have highlighted the role of specialized cellular protrusions, known as cytonemes, in mediating long-range signaling between cells, which is critical for proper tissue development[2]. Additionally, the Hippo signaling pathway has emerged as a key regulator of both cell fate and cell-cell communication, illustrating the complex interplay between signaling pathways and cellular interactions[3].

The current state of research in CCC reveals a rapidly evolving landscape. Advances in imaging techniques and molecular biology have provided new insights into the dynamic nature of cell interactions and the signaling pathways that govern them. Techniques such as microfluidics are enabling researchers to replicate the cellular microenvironment and study CCC in real-time, further elucidating the complexities of intercellular communication[4]. Furthermore, the identification of novel signaling molecules and the exploration of bioelectrical controls in tissue dynamics have opened new avenues for understanding how cells communicate and coordinate their functions[5].

This review will systematically explore the mechanisms of cell-cell communication, emphasizing their roles in coordinating tissue function. The report is organized as follows: Section 2 will detail the various mechanisms of CCC, including direct cell-cell interactions, paracrine signaling, and the role of extracellular vesicles. Section 3 will discuss the critical functions of CCC in tissue development, homeostasis, and the coordination of cellular responses. Section 4 will delve into the dysregulation of CCC in diseases such as cancer, inflammatory conditions, and neurodegenerative disorders, highlighting the pathological implications of disrupted communication pathways. Section 5 will focus on the therapeutic implications of targeting these communication pathways, including strategies for cancer therapy and tissue regeneration. Finally, Section 6 will outline future directions in research, emphasizing the need for integrative approaches to further our understanding of tissue dynamics.

By synthesizing current research findings, this review aims to provide comprehensive insights into how cell-cell interactions influence tissue dynamics and to highlight potential therapeutic strategies targeting these pathways for disease intervention. Understanding the complexities of CCC not only enhances our knowledge of fundamental biological processes but also paves the way for innovative approaches in precision medicine and regenerative therapies[1][3].

2 Mechanisms of Cell-Cell Communication

2.1 Direct Cell-Cell Interactions

Cell-cell communication is integral to the coordination of tissue function, primarily facilitated through direct interactions between cells. This communication is crucial for various cellular processes, including proliferation, survival, differentiation, and maintaining the structural integrity of tissues. Direct contact between cells enables them to exchange signals and respond to their environment effectively.

One significant mechanism of direct cell-cell interaction involves the use of specialized structures such as gap junctions, which allow the passage of small molecules and ions between adjacent cells. These junctions enable coordinated responses among cells in a tissue, facilitating rapid communication essential for tissue homeostasis. For instance, cells can synchronize their activities in response to stimuli, which is vital in processes like cardiac function and neuronal signaling.

Additionally, cells utilize membrane receptors and ligands to communicate through physical interactions. These trans interactions activate receptors on one cell, leading to changes in the fate and function of the receptor-expressing cells. Such signaling is especially important in the immune and nervous systems, where precise communication is necessary for proper function (Hui 2023).

Cis interactions, where receptors and ligands on the same cell interact, also play a critical role in regulating cell functions. This type of communication can influence how cells respond to their environment and interact with neighboring cells, highlighting a previously underappreciated regulatory mechanism in cell biology (Hui 2023).

Moreover, tunneling nanotubes (TNTs) have emerged as another mode of direct cell-cell communication. These structures facilitate the exchange of cellular cargo, including signaling molecules and organelles, thereby allowing cells to communicate over longer distances than traditional direct contact would permit. This mechanism is particularly relevant in the brain, where it enables crosstalk between various cell types, such as neurons and glial cells, crucial for maintaining brain function and responding to pathological conditions (Khattar et al. 2022).

In summary, the coordination of tissue function through cell-cell communication relies on a complex interplay of direct interactions via gap junctions, receptor-ligand engagements, and novel structures like tunneling nanotubes. These mechanisms ensure that cells can effectively communicate, adapt to their environment, and maintain the overall integrity and functionality of tissues (Lin et al. 2023; Eugenin et al. 2022).

2.2 Paracrine Signaling

Cell-cell communication is a fundamental aspect of tissue function, with paracrine signaling serving as a crucial mechanism through which cells coordinate their activities and responses to various stimuli. Paracrine signaling involves the release of signaling molecules by one cell type that diffuse locally to affect neighboring cells, thus facilitating intercellular communication and coordination.

One of the primary roles of paracrine signaling is to mediate responses to tissue injury and regeneration. For instance, in postmitotic mammalian cells, such as neurons and cardiomyocytes, the capacity for regeneration is limited. Recent studies indicate that redox signaling through paracrine mechanisms can play a significant role in linking tissue injury with regenerative responses. This type of signaling can occur via diffusion to adjacent cells, through mitochondrial interactions, or within extracellular vesicles, ultimately influencing intracellular targets like kinases, phosphatases, and transcription factors that trigger regenerative processes (Hervera et al. 2019) [6].

Additionally, intercellular communication in tissues, such as bone, relies on paracrine stimulation and gap junctions to create complex cellular networks. In human osteoblast-like cells, for example, mechanical stimulation of one cell can initiate a wave of calcium signaling that propagates to surrounding cells, highlighting the importance of paracrine signaling in maintaining tissue homeostasis (Romanello and D'Andrea 2001) [7]. This mechanism is essential for coordinating metabolic responses and adapting to mechanical stimuli.

Paracrine signaling also plays a vital role in various physiological processes, including development and homeostasis in multicellular organisms. The anterior pituitary, for instance, relies on paracrine and autocrine loops during fetal and postnatal development, with numerous signaling molecules such as neurotransmitters, growth factors, and cytokines being identified as key players in these processes (Denef 2008) [8].

In the context of cardiac function, communication between cardiomyocytes and fibroblasts is regulated through paracrine signaling, which influences cardiac development and response to pathological conditions like ischemia-reperfusion injury. The exchange of signaling molecules between these cell types can either promote tissue protection or contribute to damage, depending on the nature and concentration of the secreted factors (Flores-Vergara et al. 2021) [9].

Furthermore, advances in methodologies for studying paracrine signaling, such as high-throughput mapping of paracrine mediators, have revealed the complexity of these interactions at the single-cell level. This research indicates that variations in secretion dynamics among different cell types can significantly influence their communication and functional outcomes (Deng et al. 2025) [10].

In summary, paracrine signaling is a pivotal mechanism for coordinating tissue function by facilitating localized communication between cells. This type of signaling is essential for regulating various physiological processes, responding to environmental changes, and maintaining tissue integrity and homeostasis. Understanding these mechanisms is crucial for developing therapeutic strategies aimed at enhancing tissue repair and regeneration.

2.3 Extracellular Vesicles and Signaling Molecules

Cell-to-cell communication is fundamental for the coordination and proper organization of different cell types within multicellular systems. This communication is facilitated through various mechanisms, including the release of signaling molecules such as growth factors and chemokines, direct cell-cell contact, and the secretion of extracellular matrix components. However, a significant advancement in our understanding of cellular communication has been the recognition of extracellular vesicles (EVs) as specialized carriers of biological information.

Extracellular vesicles are nano-sized membranous structures that originate from various cellular compartments, including exosomes, microvesicles, and apoptotic bodies. They are involved in the transfer of a diverse range of molecular cargo, including proteins, lipids, RNA, and other bioactive molecules, thus influencing the behavior of recipient cells. The biogenesis of exosomes, for instance, occurs within the endosomal compartment, leading to the formation of multivesicular bodies (MVBs) that release these vesicles into the extracellular space upon fusion with the plasma membrane. This release allows EVs to interact with neighboring cells, either by inducing specific signaling pathways or altering cellular phenotypes through the transfer of receptors or genetic material [11].

In the context of cardiovascular health, for example, extracellular vesicles play a crucial role in mediating communication among various cell types, including cardiomyocytes, endothelial cells, and immune cells. They carry specific signatures of cellular activation and injury, which makes them potential biomarkers for cardiovascular diseases. The vesicles can transfer biological information through receptor activation, proteolytic enzymes, and the delivery of genetic information, thereby actively participating in inflammatory processes and tissue responses [12]. This intricate signaling network underscores the importance of EVs in both physiological and pathological conditions.

Moreover, in the central nervous system (CNS), extracellular vesicles facilitate communication between neurons and glial cells, thereby contributing to neural development and maintenance of homeostasis. They are involved in processes such as synaptic formation and myelination, demonstrating their essential role in coordinating the functions of complex neural networks [13]. The ability of EVs to cross the blood-brain barrier further emphasizes their significance in mediating communication between the CNS and peripheral systems [14].

The mechanisms by which extracellular vesicles operate are multifaceted. They can modulate the immune response by transferring immunomodulatory signals between immune and non-immune cells, thus influencing local and systemic effects in both health and disease [15]. Additionally, the heterogeneity of EVs allows for a tailored response to various stimuli, which is crucial for maintaining tissue homeostasis and responding to pathological changes [16].

In summary, extracellular vesicles serve as pivotal mediators of intercellular communication, enabling cells to exchange critical information that coordinates tissue function. Their ability to carry and transfer diverse biological signals underscores their potential as diagnostic and therapeutic tools in various diseases, further highlighting the sophisticated nature of cell-cell communication within multicellular organisms.

3 Role of Cell-Cell Communication in Tissue Function

3.1 Tissue Development and Morphogenesis

Cell-cell communication is a fundamental process that orchestrates tissue function and development in multicellular organisms. This communication is essential for coordinating various cellular behaviors such as growth, differentiation, and the formation of complex tissue structures. The intricate interplay between cells, mediated through signaling pathways and direct interactions, plays a crucial role in morphogenesis and the overall functionality of tissues.

During tissue development, particularly in processes like organogenesis, cells must coordinate their proliferation and differentiation in response to specific signals. This coordination is facilitated by intercellular communication, which involves both direct contact between cells and signaling through soluble factors. For instance, in the context of secondary palate development in mice, interactions between epithelial and mesenchymal cells are pivotal for regulating epithelial-mesenchymal signaling, which in turn controls cell proliferation, differentiation, and tissue patterning (Hussein et al., 2025). Key signaling pathways such as WNT, BMP, and PDGF are highlighted as critical regulators of these processes, with their activity peaking at specific developmental stages, thus demonstrating the temporal dynamics of cell communication in tissue morphogenesis.

The extracellular matrix (ECM) also plays a significant role in mediating cell communication. It provides biochemical and mechanical cues that influence cell behavior and tissue organization. The ECM's structural properties can dictate how cells interact with one another and respond to external signals, thereby guiding tissue morphogenesis (Gjorevski & Nelson, 2009). This bidirectional signaling, where cells not only respond to ECM signals but also remodel the ECM, is vital for shaping the local microenvironment necessary for proper tissue development.

Moreover, specialized structures such as cytonemes, which are actin-based protrusions, have been identified as crucial for long-range signaling between cells. They facilitate the precise delivery of morphogens, ensuring accurate tissue patterning and cell differentiation (Ma et al., 2025). This mechanism underscores the importance of spatial organization in cell communication, as it enables cells to communicate effectively over distances that would otherwise be challenging to coordinate through diffusion alone.

The synchronization of cellular activities is another aspect of how cell-cell communication coordinates tissue function. For example, in epithelial tissues, mechanical forces generated by cell shape changes can be sensed by adjacent cells, triggering coordinated responses across the tissue (Richa et al., 2025). This mechanotransduction allows for collective behaviors that are essential for tissue integrity and function.

In summary, cell-cell communication is a multifaceted process that involves various signaling pathways, ECM interactions, and specialized cellular structures. These elements work together to ensure that cells can effectively coordinate their behaviors during tissue development and morphogenesis. Understanding these interactions provides valuable insights into the biological processes underlying tissue homeostasis and the implications for developmental disorders and regenerative medicine (Su et al., 2024; George & Bates, 2022).

3.2 Homeostasis and Repair Mechanisms

Cell-cell communication is a fundamental aspect of maintaining tissue function, homeostasis, and facilitating repair mechanisms. It involves a variety of interactions between cells that enable them to exchange information, coordinate responses, and ensure the integrity of the tissue environment.

The Hippo pathway is a critical regulator of tissue growth and has been shown to play significant roles in cell-cell communication. This pathway influences both cell-autonomous functions, such as cell proliferation and differentiation, and non-cell-autonomous functions, which are crucial for regulating interactions between neighboring cells. The Hippo pathway mediates communication through various mechanisms, including direct cell-cell interactions, the release of soluble factors, and the involvement of extracellular vesicles and the extracellular matrix. These interactions not only dictate the fate of individual cells but also orchestrate multicellular responses that are essential for tissue homeostasis, particularly during development and regeneration [3].

In addition to the Hippo pathway, intercellular communication mechanisms such as gap junctions, paracrine signaling, and extracellular vesicles are vital for maintaining tissue integrity. For instance, in the heart, dynamic interactions among different cell types, including cardiomyocytes and cardiac fibroblasts, are crucial for coordinating heart function and responding to stressors like ischemia. These interactions facilitate the exchange of information that can lead to compensatory structural and functional changes, which are particularly important during the progression of heart failure [17].

Furthermore, adult stem cells play a significant role in tissue homeostasis by communicating with their surrounding environment. The organization and patterning of tissues reflect their functional roles, with stem cells relying on signals from neighboring cells to maintain their regenerative capacity and tissue structure. For example, epidermal stem cells communicate through Notch signaling to regulate their functions, which is essential for skin homeostasis [18].

Endocytosis is another mechanism that contributes to tissue homeostasis by regulating the levels of growth factors and maintaining a balance between different cell types. By internalizing growth factors, cells can prevent uncontrolled proliferation, which is crucial for maintaining tissue integrity [19].

Moreover, the process of intercellular communication extends beyond local interactions; it encompasses systemic responses to stress and injury. The cellular stress and immunity cycle illustrates how tissues communicate under stress conditions to recruit immune cells for repair, emphasizing the importance of these networks in both homeostasis and pathology [20].

In summary, cell-cell communication is integral to coordinating tissue function through various mechanisms that facilitate homeostasis and repair. The interplay of signaling pathways, direct interactions, and feedback mechanisms ensures that tissues can adapt to changes and maintain their structural and functional integrity. Understanding these communication networks is essential for developing therapeutic strategies aimed at enhancing tissue repair and addressing diseases that disrupt these processes.

3.3 Coordination of Cellular Responses

Cell-cell communication (CCC) is fundamental for the coordination of tissue function in multicellular organisms. This communication is essential for various cellular processes, including growth, development, differentiation, and the maintenance of tissue integrity. CCC can occur through direct contact or via ligand-receptor interactions over distances, facilitating both signal conduction and signal transduction, which are crucial for intercellular signaling and the regulation of physiological processes[1].

The mechanisms of CCC encompass a range of processes, including the exchange of signaling molecules, such as cytokines and chemokines, and the formation of specialized structures that enable direct cell-to-cell contact. For instance, tunneling nanotubes (TNTs) are recognized as conduits for cellular communication, allowing the exchange of cellular cargo between connected cells, which is vital for maintaining brain function and coordinating responses during pathological conditions[21].

Furthermore, cell-cell interactions are not merely passive exchanges; they actively shape cellular behavior and fate. For example, trans interactions between membrane receptors and ligands on adjacent cells can lead to significant changes in the functions of the responding cells, particularly in the immune and nervous systems[22]. These interactions help orchestrate complex biological responses, enabling tissues to adapt and respond to environmental cues effectively.

In the context of tissue homeostasis, CCC plays a critical role in regulating physiological processes such as tissue repair and regeneration. Disruption of these communication pathways can lead to various diseases, underscoring the importance of maintaining effective CCC for the integrity of tissue structures[23]. Recent advancements in technologies, such as single-cell sequencing and spatial transcriptomics, have enhanced our understanding of how cells communicate, providing insights into the dynamics of CCC and its implications for health and disease[24].

Moreover, the distance over which cells can effectively communicate is determined by the kinetics of signal secretion and diffusion. Studies have indicated that a single cell can meaningfully propagate a soluble signal over an effective communication distance of approximately 250 microns, taking about 10-30 minutes for the process to occur. This highlights the temporal aspect of CCC, which is crucial for the timely coordination of cellular responses during tissue development and homeostasis[25].

In summary, cell-cell communication is integral to the coordination of tissue function, enabling the exchange of signals that govern cellular behaviors and responses. The intricate networks formed by these interactions are essential for maintaining the physiological balance within tissues, and their disruption can lead to significant pathological consequences. Understanding the complexities of CCC will provide a foundation for developing targeted therapies and interventions aimed at restoring normal tissue function in various diseases.

4 Dysregulation of Cell-Cell Communication in Disease

4.1 Cancer

Cell-cell communication is a critical aspect of maintaining tissue function, particularly through mechanisms such as the Notch signaling pathway and bioelectric regulation. In the context of cancer, this communication is often dysregulated, leading to significant implications for tumor development and progression.

The Notch signaling pathway plays a pivotal role in the communication between adjacent cells, particularly during organ development. It regulates processes involved in cell differentiation and can operate through direct interactions between Notch receptors and ligands on neighboring cells or through paracrine and endocrine signaling using soluble molecules. Dysregulation of the Notch pathway, which may arise from mutations in Notch-related genes or from signals in the tumor microenvironment, is frequently observed in cancer. Cancer cells can exploit this aberrant signaling to "educate" surrounding cells towards a pro-tumoral behavior. This is facilitated by the secretion of key cytokines from tumor cells and the activation of Notch signaling in stromal cells through direct cell-to-cell contact, which enhances the secretion of pro-tumorigenic cytokines. Such alterations in cell-cell communication significantly affect the cytokine network, ultimately influencing tumor progression and the immune response against the tumor[26].

In addition to the Notch signaling pathway, bioelectricity also plays a crucial role in coordinating tissue function. The cellular membrane potential (Vmem) is a fundamental property of cells that underlies bioelectricity. Cancer cells exhibit a distinct bioelectric state compared to healthy cells, which disrupts cellular signaling pathways. This bioelectric dysregulation is implicated in all three stages of carcinogenesis: initiation, promotion, and progression. Furthermore, the production of extracellular vesicles (EVs) is another mechanism through which cells communicate within the tumor microenvironment (TME). In cancer, the production and release of EVs are altered, contributing to the dysregulation of bioelectric states and cellular communication. This disruption can affect tumor growth and metastasis, highlighting the importance of bioelectric properties in cancer biology[27].

Overall, dysregulation of cell-cell communication in cancer, whether through the Notch signaling pathway or bioelectric changes, significantly impacts tissue organization and function, facilitating tumor development and progression. Understanding these mechanisms provides insights into potential therapeutic targets for cancer treatment, emphasizing the need to restore normal cell communication pathways to combat tumor growth and improve immune responses[20].

4.2 Inflammatory Diseases

Cell-cell communication is a fundamental process that coordinates tissue function, particularly in the context of inflammation and tissue repair. The interaction between various cell types, including immune cells, fibroblasts, and adipocytes, plays a crucial role in maintaining homeostasis and responding to injury. Dysregulation of these communication pathways can lead to pathological conditions, including inflammatory diseases.

In the skin, for example, injury-induced inflammation is traditionally attributed to keratinocytes and immune cells. However, recent studies highlight the significant involvement of non-immune cells, such as tissue-resident adipocytes and fibroblasts, in orchestrating pro-inflammatory signaling after injury. These cells actively participate in the inflammatory response, and alterations in their communication can lead to irregular inflammation, which is often associated with aberrant wound healing. Understanding the contributions of these mesenchymal cells to inflammation may reveal new therapeutic targets for regulating inflammation and promoting tissue repair (Cooper et al. 2021) [28].

Similarly, in the context of rheumatoid arthritis (RA), dysregulated cell signaling pathways, including the Janus Kinase/signal transducers and activators of transcription (JAK/STAT) and Toll-like receptor/nuclear factor kappa B (TLR/NF-κB) pathways, are implicated in the disease's pathogenesis. The crosstalk between these pathways can create a sustained loop of activation that exacerbates the inflammatory condition. Identifying these signaling interactions provides potential targets for therapeutic interventions aimed at mitigating inflammation in RA (Ibrahim and Huttunen 2021) [29].

In the liver, the innate immune response to sterile inflammation—such as that occurring in response to tissue injury and cell death—relies on effective cell-cell communication. Dysregulation of immune cell trafficking and function can lead to chronic inflammatory diseases. The role of various innate immune cells, including macrophages and platelets, is critical in coordinating tissue repair. A better understanding of these mechanisms can inform strategies to address the misdirection of sterile inflammation and promote healing (McDonald and Kubes 2016) [30].

Moreover, aging is associated with immune dysfunction and changes in inflammatory pathways, which contribute to tissue deterioration. Interventions targeting chronic inflammation and enhancing tissue repair capacity are proposed as potential anti-aging strategies. By modulating inflammatory pathways, it may be possible to rejuvenate tissue function and combat age-related diseases (Neves and Sousa-Victor 2020) [31].

The dynamic nature of the inflammatory microenvironment also plays a significant role in tissue regeneration. Inflammation is necessary for tissue repair, but when it becomes chronic, it can lead to fibrosis and other complications. Understanding the interactions among immune cells and the metabolic responses during tissue regeneration is essential for developing novel therapeutic strategies, including the use of biomaterials to enhance healing (Mata et al. 2021) [32].

In conclusion, cell-cell communication is vital for coordinating tissue function, particularly during inflammatory responses. Dysregulation of these interactions can lead to a variety of inflammatory diseases, highlighting the importance of understanding these pathways for developing effective therapeutic interventions.

4.3 Neurodegenerative Disorders

Cell-cell communication plays a pivotal role in coordinating tissue function, particularly within the central nervous system (CNS). In the context of neurodegenerative disorders, the dysregulation of such communication can significantly contribute to disease progression and pathology.

Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by a disrupted neuroinflammatory environment that stems from the dysregulation of neuroglial intercellular communication. This communication is primarily mediated by extracellular signals and the activity of connexins (Cxs), which form gap junctions (Gjs) and hemichannels (HCs). These structures are crucial for maintaining both intracellular and extracellular homeostasis. However, under pathological conditions, connexins can function as pathological pores, leading to synaptic dysfunction, oxidative stress, and ultimately cell death [33].

The communication between glial cells and neurons is essential for maintaining neuronal health and overall brain homeostasis. In Alzheimer's disease, for instance, altered glia-neuron communication has been observed, particularly in the hippocampus of 3xTg-AD mice. Studies employing single-nucleus RNA sequencing revealed that this communication becomes increasingly dysregulated over time, affecting the gene regulatory mechanisms in neurons [34]. Specifically, the study identified 23 AD-associated ligand-receptor pairs that are upregulated in the hippocampus of older 3xTg-AD mice, suggesting that microglial signaling interactions may play a significant role in the disease [34].

Moreover, dysfunctional intercellular communication is not limited to glial and neuronal interactions; it also encompasses the crosstalk between neurons and the innate immune system. Evidence indicates that non-cell-autonomous processes, such as pathological cell-cell communication, are critical in the initiation and progression of neurodegenerative diseases. This highlights the complexity of intercellular perturbations and their implications for therapeutic approaches [35].

The implications of disrupted cell-cell communication extend beyond mere signaling failures. For instance, nuclear import defects and cell cycle dysregulation have been identified as common drivers of pathology in neurodegeneration. Aberrant activation of the cell cycle in post-mitotic neurons can trigger cell death pathways, exacerbating neurodegenerative conditions [36]. Additionally, disturbances in interorganelle communication have been linked to aging and neurodegeneration, emphasizing the need for coordinated organelle function for cellular homeostasis [37].

Overall, the coordination of tissue function through cell-cell communication is crucial for maintaining neuronal integrity and function. Dysregulation of this communication in neurodegenerative disorders leads to a cascade of pathological events, including neuroinflammation, synaptic dysfunction, and neuronal loss. Understanding these mechanisms is vital for developing targeted therapeutic strategies aimed at restoring normal intercellular communication and mitigating the effects of neurodegenerative diseases.

5 Therapeutic Implications

5.1 Targeting Communication Pathways in Cancer Therapy

Cell-cell communication plays a critical role in coordinating tissue function, particularly in the context of cancer. The interactions between cells within a tissue can dictate various physiological processes, and when these communication pathways are disrupted, it can lead to pathological conditions such as cancer. In cancer, cellular communication is not limited to interactions between cancer cells themselves but also includes communication with immune cells, neural cells, and other components of the tumor microenvironment.

One of the mechanisms of cell-cell communication is bioelectric signaling, which involves the exchange of ions and the generation of electric fields that influence cellular behavior. This type of signaling has been shown to affect tumor growth and invasion, suggesting that alterations in bioelectric properties can contribute to cancer progression (McMillen et al., 2021) [38]. Additionally, tunneling nanotubes (TNTs) facilitate the transport of molecules between cells, enabling long-distance signaling that can impact tissue homeostasis and malignancy (McMillen et al., 2021) [38].

Moreover, the immune system plays a pivotal role in the communication network within tumors. Recent studies have highlighted that the nervous system and immune cells communicate to regulate inflammation and host responses, which is crucial in cancer initiation and progression. For instance, sympathetic nerve activity can influence tumor microenvironments by modulating immune cell functions, potentially promoting or inhibiting tumor growth depending on the tumor type (Scheff & Saloman, 2021) [39].

The implications for therapy are profound. Targeting these communication pathways presents novel therapeutic opportunities. For instance, metabolic reprogramming within the tumor microenvironment can influence anti-tumor immunity. By understanding how metabolic processes affect immune cell functions, strategies can be developed to enhance the efficacy of cancer immunotherapies (Chapman & Chi, 2024) [40]. Additionally, interventions aimed at modifying neural activity, such as suppressing sympathetic activity or enhancing parasympathetic responses, have been shown to activate anti-tumor immune responses while alleviating cancer-associated symptoms (Scheff & Saloman, 2021) [39].

Furthermore, the interplay between various signaling pathways, such as the Hippo signaling pathway and others like Wnt or TGF-β, illustrates the complexity of communication in cancer. Understanding these interactions could lead to targeted therapies that exploit specific vulnerabilities in cancer cell signaling (Noorbakhsh et al., 2021) [41].

In summary, the coordination of tissue function through cell-cell communication is vital for maintaining homeostasis and regulating responses to cancer. By targeting these communication pathways, particularly those involving metabolic processes, neural activity, and key signaling networks, there is potential to develop innovative therapeutic strategies that not only suppress tumor growth but also mitigate the neurological and immune-related complications associated with cancer. This holistic approach may significantly enhance the effectiveness of current cancer therapies and improve patient outcomes.

5.2 Modulating Cell-Cell Interactions for Tissue Regeneration

Cell-cell communication is a fundamental process that underpins the coordinated function of tissues in multicellular organisms. This communication can occur through various modalities, including direct contact, paracrine signaling, and the exchange of extracellular vesicles (EVs). The mechanisms by which cells communicate are essential for regulating key cellular processes such as proliferation, differentiation, and survival, and they play a crucial role in maintaining tissue homeostasis.

The extracellular matrix (ECM) serves as a critical mediator of cell-cell communication, providing structural support while actively modulating cellular responses. In both physiological and pathological contexts, particularly in cancer, ECM reorganization influences tumor development and progression by shaping interactions within the tumor microenvironment (TME) [42]. The biomechanical properties of the ECM, along with the controlled release of EVs, facilitate the transmission of signaling molecules, proteins, and microRNAs that modulate cellular behavior and can initiate processes such as metastasis [42].

In the context of tissue regeneration, cell-cell communication is vital for orchestrating the repair and regeneration processes. Research has shown that cytokines play a pivotal role in intercellular communication during skeletal muscle regeneration, where resident muscle stem cells communicate extensively with the microenvironment to coordinate repair efforts [43]. The dynamic expression of cytokines by various cell types at the injury site facilitates bidirectional communication, guiding the regeneration process and ensuring successful tissue repair [43].

Therapeutically, the modulation of cell-cell interactions presents significant opportunities for enhancing tissue regeneration. Targeting ECM dynamics, for instance, could improve therapeutic outcomes by optimizing the signaling pathways involved in cell communication. Recent advancements in regenerative medicine highlight the potential of manipulating bioelectrical signals and biophysical properties of tissues to influence cellular behaviors and promote regeneration [5], [44]. Such approaches could enable the development of novel therapeutic strategies that harness the inherent regenerative capacities of tissues, particularly in contexts where natural repair mechanisms are insufficient, such as in chronic injuries or degenerative diseases.

Moreover, understanding the mechanisms of cell competition—where more competitive cells can eliminate less fit neighbors—provides insights into the regulatory frameworks governing tissue growth and regeneration [45]. This knowledge could inform therapeutic interventions aimed at enhancing the regenerative potential of stem cells or improving the efficacy of cell-based therapies.

In conclusion, the intricate network of cell-cell communication is central to coordinating tissue function and regeneration. By leveraging insights into these communication pathways, particularly those involving the ECM, cytokines, and bioelectrical signals, therapeutic strategies can be developed to enhance tissue repair and regeneration, offering promising avenues for addressing various medical challenges in regenerative medicine.

5.3 Innovations in Drug Delivery via Extracellular Vesicles

Cell-cell communication is a fundamental aspect of maintaining tissue homeostasis and coordinating physiological functions across multicellular organisms. This intricate communication network is facilitated by various mechanisms, one of which involves extracellular vesicles (EVs). These membrane-bound structures, including exosomes and microvesicles, play a crucial role in transmitting biological information between cells, influencing processes such as cellular differentiation, proliferation, and immune responses.

Extracellular vesicles are released from virtually all cell types and carry diverse cargos, including proteins, lipids, and nucleic acids, which can modulate the behavior of recipient cells. For instance, in the context of cardiovascular diseases, EVs can transfer signaling molecules that influence cardiac function and structure. Under pathological conditions, the composition of these vesicles changes, contributing to disease progression. Investigating the molecular cargo of EVs is essential for understanding their role in tissue function and disease mechanisms [16][46].

The therapeutic implications of EVs are vast, particularly in the development of novel drug delivery systems. Their unique properties, such as biocompatibility, stability, and low immunogenicity, make them promising candidates for delivering therapeutic agents. Engineered EVs can be tailored to enhance drug targeting and release, providing a "cell-free, cell therapy" approach for treating various conditions, including cancer and cardiovascular diseases [47][48]. This innovative strategy leverages the natural ability of EVs to communicate and deliver biological information, thereby opening new avenues for therapeutic interventions [49].

Recent advances in understanding EV biology have highlighted their potential as biomarkers for disease diagnosis and prognosis. Their ability to encapsulate specific molecular signatures reflective of the pathological state of tissues makes them valuable tools for non-invasive diagnostics [50][51]. As research continues to unravel the complexities of EV-mediated signaling, the potential for developing effective therapeutic strategies that harness these vesicles for drug delivery and disease management becomes increasingly viable [52][53].

In summary, the coordination of tissue function through cell-cell communication via extracellular vesicles is a dynamic and complex process with significant therapeutic implications. Innovations in drug delivery utilizing EVs represent a promising frontier in biomedical research, potentially transforming the landscape of treatment for various diseases.

6 Future Directions in Research

6.1 Advances in Imaging Techniques

Cell-cell communication (CCC) is a fundamental process that coordinates the functions of tissues in multicellular organisms. This coordination is achieved through various mechanisms, including direct contact and the use of signaling molecules, which facilitate interactions between different cell types. The significance of CCC in maintaining tissue homeostasis, regulating development, and orchestrating immune responses cannot be overstated.

Research has shown that cells communicate through ligand-receptor interactions, which are critical for signal conduction and transduction. These processes involve the generation and transmission of signals between cells, allowing for coordinated responses essential for growth, differentiation, and physiological regulation [1]. Furthermore, understanding the complexities of these interactions is vital for deciphering the mechanisms underlying tissue organization and function, particularly in the context of diseases where communication pathways may be disrupted.

Advancements in imaging techniques have significantly enhanced our ability to study CCC. For instance, microfluidic devices have emerged as powerful tools that allow researchers to manipulate and observe cell interactions in a highly controlled environment. These devices facilitate both short-distance (single-cell) and long-distance (population-level) communication studies, enabling high-resolution imaging and real-time analysis of cellular dynamics [54]. The integration of sensors within these platforms further augments the ability to monitor signaling events and cellular responses, thus providing deeper insights into the regulatory pathways involved in tissue function.

In addition to imaging advancements, the exploration of cis interactions between membrane receptors and ligands presents a new dimension in understanding CCC. Unlike traditional trans signaling, which focuses on interactions between different cells, cis interactions occur between receptors and ligands on the same cell, potentially influencing cell behavior and function in novel ways [22]. This area of research highlights the complexity of cellular communication networks and their implications for tissue homeostasis and pathology.

Moreover, bioelectrical signaling has been identified as a critical component of CCC, particularly in the context of morphogenesis and tissue regeneration. The propagation of electrical signals across cell membranes facilitates rapid and efficient communication, allowing for coordinated cellular responses that are essential for tissue development and repair [5]. Understanding how these bioelectrical signals interact with biochemical pathways opens new avenues for therapeutic interventions in regenerative medicine.

In conclusion, the coordination of tissue function through cell-cell communication is a multifaceted process that involves various signaling mechanisms and advanced imaging techniques. Future research is poised to explore these interactions in greater detail, leveraging new technologies to uncover the intricacies of cellular communication and its impact on health and disease. This comprehensive understanding is essential for the development of targeted therapies and personalized medicine approaches aimed at restoring normal tissue function.

6.2 Exploring Novel Signaling Molecules

Cell-cell communication (CCC) plays a pivotal role in coordinating tissue function, particularly in multicellular organisms where diverse cell types must interact harmoniously for proper physiological processes. This communication is essential for various functions including growth, development, differentiation, and maintenance of tissues and organs. Cells communicate through direct contact or via signaling molecules, utilizing ligand-receptor interactions to transmit information both locally and over distances. The processes involved in CCC include cell signal conduction, which generates and transmits signals, and signal transduction, which involves the reception and processing of these signals [1].

Understanding the mechanisms of CCC is crucial for elucidating how cellular interactions influence biological functions. Recent advancements in imaging technologies have enhanced the identification of cellular protrusions, such as cilia and microvilli, which are specialized structures facilitating selective and efficient signaling between cells. The regulation of these protrusions is vital for maintaining appropriate signaling outcomes, as they enable cells to establish connections and communicate effectively within complex tissues [55].

Moreover, the discovery of novel signaling pathways, such as the transport of macromolecules through plasmodesmata in plants, highlights the diversity of communication mechanisms across different organisms. This transport system allows for the exchange of proteins and nucleic acids, suggesting a sophisticated form of cell-to-cell signaling that can influence developmental processes [56]. Additionally, the role of gap junctions in enabling the direct passage of small signaling molecules between adjacent cells underscores the importance of such structures in coordinating tissue activity [57].

As research progresses, it is essential to explore new signaling molecules and pathways that may further elucidate the complexity of CCC. Investigating the bioelectrical signals that govern cellular behaviors can provide insights into how cells coordinate responses to their microenvironment, particularly in developmental biology and regenerative medicine [5]. Furthermore, understanding the role of cis interactions between membrane receptors and ligands could unveil additional regulatory mechanisms that impact immune and other cellular functions [22].

Future research directions should focus on the integration of these novel signaling molecules into existing frameworks of cellular communication. This involves not only identifying new players in CCC but also understanding their interactions and effects on tissue homeostasis and pathology. By enhancing our comprehension of these complex networks, we can develop targeted therapeutic strategies that address diseases resulting from communication breakdowns, thus advancing the field of precision medicine [1].

6.3 Integrative Approaches to Study Tissue Dynamics

Cell-to-cell communication is fundamental for the coordination of tissue function in multicellular organisms. This communication facilitates the sharing of information between cells through various mechanisms, including biochemical signaling, electrical signals, and physical interactions between receptors and ligands.

One of the primary modes of intercellular communication involves the release of soluble cytokines and chemokines, which diffuse through the extracellular medium and bind to receptors on neighboring cells. This process is governed by both physicochemical transport processes and cellular secretion rates, which are influenced by genetic and biochemical factors. Effective communication distances for these soluble signals can reach approximately 250 microns, with the signaling process taking between 10 to 30 minutes to propagate meaningfully (Francis and Palsson, 1997) [25].

In addition to soluble factors, bioelectrical signals play a crucial role in coordinating cellular behaviors and decisions. Recent advancements in understanding how cells utilize bioelectrical signals have revealed their importance in developmental biology and regenerative medicine. Researchers are now able to predictively alter these signals to orchestrate complex pattern formation and tissue regeneration, indicating a promising avenue for therapeutic applications (Whited and Levin, 2019) [5].

Furthermore, the regulation of intercellular communication is intricately linked to the actin cytoskeleton and the transforming growth factor β (TGFβ) signaling pathway. The actin cytoskeleton, connected to the extracellular matrix through integrins, creates a platform for biochemical and biomechanical signaling that is crucial for maintaining tissue homeostasis. Disruptions in these signaling pathways can lead to pathological conditions, including cancer, highlighting the importance of understanding these interactions (Melchionna et al., 2021) [58].

As research progresses, it is essential to adopt integrative approaches that combine quantitative analysis of intercellular signaling with studies of tissue architecture and dynamics. This includes exploring the competitive and cooperative interactions among cells that shape tissue organization during homeostasis and tumorigenesis. By employing models that capture these interactions, researchers can gain insights into the evolutionary dynamics of tissue architecture and its implications for health and disease (Csikász-Nagy et al., 2013) [59].

Future directions in this field may involve deeper investigations into the cis interactions between membrane receptors and ligands, which have been shown to regulate immune cell functions and other cellular processes. Understanding these understudied regulatory mechanisms could provide new insights into the complex web of cell communication and its impact on tissue function (Hui, 2023) [22].

In summary, the coordination of tissue function through cell-cell communication is a multifaceted process involving biochemical signals, bioelectrical changes, and physical interactions, all of which warrant further exploration to uncover their roles in health and disease.

7 Conclusion

The review highlights the critical role of cell-cell communication (CCC) in coordinating tissue function across multicellular organisms. Key findings reveal that various mechanisms, including direct interactions through gap junctions, paracrine signaling, and the involvement of extracellular vesicles, are fundamental for maintaining tissue homeostasis, development, and repair. The exploration of novel structures like tunneling nanotubes and specialized cellular protrusions, such as cytonemes, underscores the complexity of CCC and its importance in facilitating long-range signaling and precise tissue patterning. Dysregulation of these communication pathways is implicated in numerous diseases, including cancer, inflammatory disorders, and neurodegenerative conditions, emphasizing the need for a deeper understanding of CCC in health and disease contexts. Future research should focus on integrative approaches that combine advanced imaging techniques, the identification of novel signaling molecules, and the exploration of bioelectrical signaling to further elucidate the dynamics of CCC. This understanding is essential for developing targeted therapeutic strategies aimed at restoring normal tissue function and enhancing regenerative processes, ultimately paving the way for innovations in precision medicine and regenerative therapies.

References

  • [1] Jimeng Su;Ying Song;Zhipeng Zhu;Xinyue Huang;Jibiao Fan;Jie Qiao;Fengbiao Mao. Cell-cell communication: new insights and clinical implications.. Signal transduction and targeted therapy(IF=52.7). 2024. PMID:39107318. DOI: 10.1038/s41392-024-01888-z.
  • [2] Chengting Zhang;Steffen Scholpp. Cytonemes in development.. Current opinion in genetics & development(IF=3.6). 2019. PMID:31404787. DOI: 10.1016/j.gde.2019.06.005.
  • [3] Akihiro Nita;Toshiro Moroishi. Hippo pathway in cell-cell communication: emerging roles in development and regeneration.. Inflammation and regeneration(IF=6.6). 2024. PMID:38566194. DOI: 10.1186/s41232-024-00331-8.
  • [4] Feng Guo;Jarrod B French;Peng Li;Hong Zhao;Chung Yu Chan;James R Fick;Stephen J Benkovic;Tony Jun Huang. Probing cell-cell communication with microfluidic devices.. Lab on a chip(IF=5.4). 2013. PMID:23843092. DOI: 10.1039/c3lc90067c.
  • [5] Jessica L Whited;Michael Levin. Bioelectrical controls of morphogenesis: from ancient mechanisms of cell coordination to biomedical opportunities.. Current opinion in genetics & development(IF=3.6). 2019. PMID:31442749. DOI: 10.1016/j.gde.2019.06.014.
  • [6] Arnau Hervera;Celio X Santos;Francesco De Virgiliis;Ajay M Shah;Simone Di Giovanni. Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration.. Trends in cell biology(IF=18.1). 2019. PMID:30795898. DOI: 10.1016/j.tcb.2019.01.006.
  • [7] M Romanello;P D'Andrea. Dual mechanism of intercellular communication in HOBIT osteoblastic cells: a role for gap-junctional hemichannels.. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research(IF=5.9). 2001. PMID:11499869. DOI: 10.1359/jbmr.2001.16.8.1465.
  • [8] C Denef. Paracrinicity: the story of 30 years of cellular pituitary crosstalk.. Journal of neuroendocrinology(IF=4.1). 2008. PMID:18081553. DOI: 10.1111/j.1365-2826.2007.01616.x.
  • [9] Raúl Flores-Vergara;Ivonne Olmedo;Pablo Aránguiz;Jaime Andrés Riquelme;Raúl Vivar;Zully Pedrozo. Communication Between Cardiomyocytes and Fibroblasts During Cardiac Ischemia/Reperfusion and Remodeling: Roles of TGF-β, CTGF, the Renin Angiotensin Axis, and Non-coding RNA Molecules.. Frontiers in physiology(IF=3.4). 2021. PMID:34539441. DOI: 10.3389/fphys.2021.716721.
  • [10] Jiu Deng;Shuai Yuan;Shangwen Yang;Zhijiang Zhang;Quanfeng Deng;Chengjun Wu. Dissecting Cellular Diversity in Paracrine Signaling with Single-Cell Secretion Profiling.. ACS sensors(IF=9.1). 2025. PMID:40679837. DOI: 10.1021/acssensors.5c00845.
  • [11] Chiara Corrado;Stefania Raimondo;Antonio Chiesi;Francesco Ciccia;Giacomo De Leo;Riccardo Alessandro. Exosomes as intercellular signaling organelles involved in health and disease: basic science and clinical applications.. International journal of molecular sciences(IF=4.9). 2013. PMID:23466882. DOI: 10.3390/ijms14035338.
  • [12] Xavier Loyer;Anne-Clémence Vion;Alain Tedgui;Chantal M Boulanger. Microvesicles as cell-cell messengers in cardiovascular diseases.. Circulation research(IF=16.2). 2014. PMID:24436430. DOI: 10.1161/CIRCRESAHA.113.300858.
  • [13] Nasim Bahram Sangani;Ana Rita Gomes;Leopold M G Curfs;Chris P Reutelingsperger. The role of Extracellular Vesicles during CNS development.. Progress in neurobiology(IF=6.1). 2021. PMID:34314775. DOI: 10.1016/j.pneurobio.2021.102124.
  • [14] Francesca Martina Filannino;Maria Antonietta Panaro;Tarek Benameur;Ilaria Pizzolorusso;Chiara Porro. Extracellular Vesicles in the Central Nervous System: A Novel Mechanism of Neuronal Cell Communication.. International journal of molecular sciences(IF=4.9). 2024. PMID:38338906. DOI: 10.3390/ijms25031629.
  • [15] Priya Makhijani;Tracy L McGaha. Myeloid Responses to Extracellular Vesicles in Health and Disease.. Frontiers in immunology(IF=5.9). 2022. PMID:35320943. DOI: 10.3389/fimmu.2022.818538.
  • [16] Junjie Xiao;Joost P G Sluijter. Extracellular vesicles in cardiovascular homeostasis and disease: potential role in diagnosis and therapy.. Nature reviews. Cardiology(IF=44.2). 2025. PMID:40045042. DOI: 10.1038/s41569-025-01141-2.
  • [17] Claudia Bang;Charalambos Antoniades;Alexios S Antonopoulos;Ulf Eriksson;Constantijn Franssen;Nazha Hamdani;Lorenz Lehmann;Christine Moessinger;Marco Mongillo;Lars Muhl;Thimoteus Speer;Thomas Thum. Intercellular communication lessons in heart failure.. European journal of heart failure(IF=10.8). 2015. PMID:26398116. DOI: 10.1002/ejhf.399.
  • [18] Valerie Horsley. Lifting Each Other Up: Epidermal Stem Cells in Tissue Homeostasis.. Developmental cell(IF=8.7). 2019. PMID:31689385. DOI: 10.1016/j.devcel.2019.10.013.
  • [19] Miri Adler;Avi Mayo;Xu Zhou;Ruth A Franklin;Jeremy B Jacox;Ruslan Medzhitov;Uri Alon. Endocytosis as a stabilizing mechanism for tissue homeostasis.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2018. PMID:29429964. DOI: 10.1073/pnas.1714377115.
  • [20] Raquel S Laureano;Isaure Vanmeerbeek;Jenny Sprooten;Jannes Govaerts;Stefan Naulaerts;Abhishek D Garg. The cell stress and immunity cycle in cancer: Toward next generation of cancer immunotherapy.. Immunological reviews(IF=8.3). 2024. PMID:37937803. DOI: 10.1111/imr.13287.
  • [21] Khattar E Khattar;Janice Safi;Anne-Marie Rodriguez;Marie-Luce Vignais. Intercellular Communication in the Brain through Tunneling Nanotubes.. Cancers(IF=4.4). 2022. PMID:35267518. DOI: 10.3390/cancers14051207.
  • [22] Enfu Hui. Cis Interactions of Membrane Receptors and Ligands.. Annual review of cell and developmental biology(IF=11.4). 2023. PMID:37339682. DOI: 10.1146/annurev-cellbio-120420-103941.
  • [23] Ling Lin;Yandong Zhou;Kebin Hu. Cell-Cell Communication and Extracellular Vesicles in Cancer.. Cancers(IF=4.4). 2023. PMID:37173886. DOI: 10.3390/cancers15092419.
  • [24] Hyobin Kim;Kwang-Eun Kim;Esha Madan;Patrick Martin;Rajan Gogna;Hyun-Woo Rhee;Kyoung-Jae Won. Unveiling contact-mediated cellular crosstalk.. Trends in genetics : TIG(IF=16.3). 2024. PMID:38906738. DOI: 10.1016/j.tig.2024.05.010.
  • [25] K Francis;B O Palsson. Effective intercellular communication distances are determined by the relative time constants for cyto/chemokine secretion and diffusion.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 1997. PMID:9356436. DOI: 10.1073/pnas.94.23.12258.
  • [26] Michela Colombo;Leonardo Mirandola;Maurizio Chiriva-Internati;Andrea Basile;Massimo Locati;Elena Lesma;Raffaella Chiaramonte;Natalia Platonova. Cancer Cells Exploit Notch Signaling to Redefine a Supportive Cytokine Milieu.. Frontiers in immunology(IF=5.9). 2018. PMID:30154786. DOI: 10.3389/fimmu.2018.01823.
  • [27] Maulee Sheth;Leyla Esfandiari. Bioelectric Dysregulation in Cancer Initiation, Promotion, and Progression.. Frontiers in oncology(IF=3.3). 2022. PMID:35359398. DOI: 10.3389/fonc.2022.846917.
  • [28] Paula O Cooper;MaryEllen R Haas;Satish Kumar R Noonepalle;Brett A Shook. Dermal Drivers of Injury-Induced Inflammation: Contribution of Adipocytes and Fibroblasts.. International journal of molecular sciences(IF=4.9). 2021. PMID:33669239. DOI: 10.3390/ijms22041933.
  • [29] Sherihan Salaheldin Abdelhamid Ibrahim;Kristiina M Huttunen. Orchestrated modulation of rheumatoid arthritis via crosstalking intracellular signaling pathways.. Inflammopharmacology(IF=5.3). 2021. PMID:33740220. DOI: 10.1007/s10787-021-00800-3.
  • [30] Braedon McDonald;Paul Kubes. Innate Immune Cell Trafficking and Function During Sterile Inflammation of the Liver.. Gastroenterology(IF=25.1). 2016. PMID:27725145. DOI: 10.1053/j.gastro.2016.09.048.
  • [31] Joana Neves;Pedro Sousa-Victor. Regulation of inflammation as an anti-aging intervention.. The FEBS journal(IF=4.2). 2020. PMID:31529582. DOI: 10.1111/febs.15061.
  • [32] Rani Mata;Yuejun Yao;Wangbei Cao;Jie Ding;Tong Zhou;Zihe Zhai;Changyou Gao. The Dynamic Inflammatory Tissue Microenvironment: Signality and Disease Therapy by Biomaterials.. Research (Washington, D.C.)(IF=10.7). 2021. PMID:33623917. DOI: 10.34133/2021/4189516.
  • [33] Simona Denaro;Simona D'Aprile;Nunzio Vicario;Rosalba Parenti. Mechanistic insights into connexin-mediated neuroglia crosstalk in neurodegenerative diseases.. Frontiers in cellular neuroscience(IF=4.0). 2025. PMID:40007760. DOI: 10.3389/fncel.2025.1532960.
  • [34] Tabea M Soelter;Timothy C Howton;Elizabeth J Wilk;Jordan H Whitlock;Amanda D Clark;Allison Birnbaum;Dalton C Patterson;Constanza J Cortes;Brittany N Lasseigne. Evaluation of altered cell-cell communication between glia and neurons in the hippocampus of 3xTg-AD mice at two time points.. Journal of cell communication and signaling(IF=3.9). 2025. PMID:40026671. DOI: 10.1002/ccs3.70006.
  • [35] Gwenn A Garden;Albert R La Spada. Intercellular (mis)communication in neurodegenerative disease.. Neuron(IF=15.0). 2012. PMID:22405200. DOI: 10.1016/j.neuron.2012.02.017.
  • [36] Jonathan Plessis-Belair;Taylor Russo;Markus Riessland;Roger B Sher. Nuclear Import Defects Drive Cell Cycle Dysregulation in Neurodegeneration.. Aging cell(IF=7.1). 2025. PMID:40377023. DOI: 10.1111/acel.70091.
  • [37] Maja Petkovic;Caitlin E O'Brien;Yuh Nung Jan. Interorganelle communication, aging, and neurodegeneration.. Genes & development(IF=7.7). 2021. PMID:33861720. DOI: 10.1101/gad.346759.120.
  • [38] Patrick McMillen;Madeleine J Oudin;Michael Levin;Samantha L Payne. Beyond Neurons: Long Distance Communication in Development and Cancer.. Frontiers in cell and developmental biology(IF=4.3). 2021. PMID:34621752. DOI: 10.3389/fcell.2021.739024.
  • [39] Nicole N Scheff;Jami L Saloman. Neuroimmunology of cancer and associated symptomology.. Immunology and cell biology(IF=3.0). 2021. PMID:34355434. DOI: 10.1111/imcb.12496.
  • [40] Nicole M Chapman;Hongbo Chi. Metabolic rewiring and communication in cancer immunity.. Cell chemical biology(IF=7.2). 2024. PMID:38428418. DOI: 10.1016/j.chembiol.2024.02.001.
  • [41] Negar Noorbakhsh;Bentolhoda Hayatmoghadam;Marzieh Jamali;Maryam Golmohammadi;Maria Kavianpour. The Hippo signaling pathway in leukemia: function, interaction, and carcinogenesis.. Cancer cell international(IF=6.0). 2021. PMID:34953494. DOI: 10.1186/s12935-021-02408-7.
  • [42] Zoi Piperigkou;Sylvia Mangani;Nikolaos E Koletsis;Christos Koutsakis;Nicholas S Mastronikolis;Marco Franchi;Nikos K Karamanos. Principal mechanisms of extracellular matrix-mediated cell-cell communication in physiological and tumor microenvironments.. The FEBS journal(IF=4.2). 2025. PMID:40747564. DOI: 10.1111/febs.70207.
  • [43] Pallob Barai;Jie Chen. Cytokine expression and cytokine-mediated cell-cell communication during skeletal muscle regeneration revealed by integrative analysis of single-cell RNA sequencing data.. Journal of cell communication and signaling(IF=3.9). 2024. PMID:39691872. DOI: 10.1002/ccs3.12055.
  • [44] Nirosha J Murugan;Solsa Cariba;Sawith Abeygunawardena;Nicolas Rouleau;Samantha L Payne. Biophysical control of plasticity and patterning in regeneration and cancer.. Cellular and molecular life sciences : CMLS(IF=6.2). 2023. PMID:38099951. DOI: 10.1007/s00018-023-05054-6.
  • [45] Rajan Gogna;Kevin Shee;Eduardo Moreno. Cell Competition During Growth and Regeneration.. Annual review of genetics(IF=8.6). 2015. PMID:26631518. DOI: 10.1146/annurev-genet-112414-055214.
  • [46] Sheila Caño-Carrillo;Juan Manuel Castillo-Casas;Diego Franco;Estefanía Lozano-Velasco. Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases.. Cells(IF=5.2). 2024. PMID:38334657. DOI: 10.3390/cells13030265.
  • [47] Kyle I Mentkowski;Jonathan D Snitzer;Sarah Rusnak;Jennifer K Lang. Therapeutic Potential of Engineered Extracellular Vesicles.. The AAPS journal(IF=3.7). 2018. PMID:29546642. DOI: 10.1208/s12248-018-0211-z.
  • [48] Inge Katrin Herrmann;Matthew John Andrew Wood;Gregor Fuhrmann. Extracellular vesicles as a next-generation drug delivery platform.. Nature nanotechnology(IF=34.9). 2021. PMID:34211166. DOI: 10.1038/s41565-021-00931-2.
  • [49] Samir EL Andaloussi;Imre Mäger;Xandra O Breakefield;Matthew J A Wood. Extracellular vesicles: biology and emerging therapeutic opportunities.. Nature reviews. Drug discovery(IF=101.8). 2013. PMID:23584393. DOI: 10.1038/nrd3978.
  • [50] Gregor Fuhrmann;Inge K Herrmann;Molly M Stevens. Cell-derived vesicles for drug therapy and diagnostics: opportunities and challenges.. Nano today(IF=10.9). 2015. PMID:28458718. DOI: 10.1016/j.nantod.2015.04.004.
  • [51] Pasquale Simeone;Giuseppina Bologna;Paola Lanuti;Laura Pierdomenico;Maria Teresa Guagnano;Damiana Pieragostino;Piero Del Boccio;Daniele Vergara;Marco Marchisio;Sebastiano Miscia;Renato Mariani-Costantini. Extracellular Vesicles as Signaling Mediators and Disease Biomarkers across Biological Barriers.. International journal of molecular sciences(IF=4.9). 2020. PMID:32260425. DOI: 10.3390/ijms21072514.
  • [52] Carlos González-Blanco;Sarai Iglesias-Fortes;Ángela Cristina Lockwood;César Figaredo;Daniela Vitulli;Carlos Guillén. The Role of Extracellular Vesicles in Metabolic Diseases.. Biomedicines(IF=3.9). 2024. PMID:38790954. DOI: 10.3390/biomedicines12050992.
  • [53] Estera Bakinowska;Kajetan Kiełbowski;Andrzej Pawlik. The Role of Extracellular Vesicles in the Pathogenesis and Treatment of Rheumatoid Arthritis and Osteoarthritis.. Cells(IF=5.2). 2023. PMID:38067147. DOI: 10.3390/cells12232716.
  • [54] Timothy Quang Vu;Ricardo Miguel Bessa de Castro;Lidong Qin. Bridging the gap: microfluidic devices for short and long distance cell-cell communication.. Lab on a chip(IF=5.4). 2017. PMID:28205652. DOI: 10.1039/c6lc01367h.
  • [55] Mayu Inaba;Sharif M Ridwan;Matthew Antel. Removal of cellular protrusions.. Seminars in cell & developmental biology(IF=6.0). 2022. PMID:35260295. DOI: 10.1016/j.semcdb.2022.02.025.
  • [56] B Ding. Cell-to-cell transport of macromolecules through plasmodesmata: a novel signalling pathway in plants.. Trends in cell biology(IF=18.1). 1997. PMID:17708891. DOI: 10.1016/S0962-8924(97)20041-3.
  • [57] Iraj Laffafian;Maurice B Hallett. Lipid-protein cargo transfer: a mode of direct cell-to-cell communication for lipids and their associated proteins.. Journal of cellular physiology(IF=4.0). 2007. PMID:17096382. DOI: 10.1002/jcp.20851.
  • [58] Roberta Melchionna;Paola Trono;Annalisa Tocci;Paola Nisticò. Actin Cytoskeleton and Regulation of TGFβ Signaling: Exploring Their Links.. Biomolecules(IF=4.8). 2021. PMID:33672325. DOI: 10.3390/biom11020336.
  • [59] Attila Csikász-Nagy;Luis M Escudero;Martial Guillaud;Sean Sedwards;Buzz Baum;Matteo Cavaliere. Cooperation and competition in the dynamics of tissue architecture during homeostasis and tumorigenesis.. Seminars in cancer biology(IF=15.7). 2013. PMID:23751796. DOI: .

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

cell-cell communication · tissue function · signaling pathways · tissue development · disease modulation


© 2025 MaltSci