Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 May 26.
Published in final edited form as: Circ Res. 2023 May 25;132(11):1546–1565. doi: 10.1161/CIRCRESAHA.123.322791

Cardiovascular Brain Circuits

Sarajo K Mohanta 1,2,§, Changjun Yin 1,3, Christian Weber 1,2, Cristina Godinho-Silva 4, Henrique Veiga-Fernandes 4, Qian J Xu 5,§, Rui B Chang 5,§, Andreas JR Habenicht 1,2,§
PMCID: PMC10231443  NIHMSID: NIHMS1895916  PMID: 37228235

Abstract

The cardiovascular system is hardwired to the brain via multilayered afferent and efferent polysynaptic axonal connections. Two major anatomically and functionally distinct though closely interacting subcircuits within the cardiovascular system have recently been defined: The artery-brain circuit and the heart-brain circuit. However, how the nervous system impacts cardiovascular disease progression remains poorly understood. Here, we review recent findings on the anatomy, structures, and inner workings of the lesser-known artery-brain circuit and the better-established heart-brain circuit. We explore the evidence that signals from arteries or the heart form a systemic and finely tuned cardiovascular brain circuit: afferent inputs originating in the arterial tree or the heart are conveyed to distinct sensory neurons in the brain. There, primary integration centers act as hubs that receive and integrate artery-brain circuit-derived and heart-brain circuit-derived signals and process them together with axonal connections and humoral cues from distant brain regions. To conclude the cardiovascular brain circuit, integration centers transmit the constantly modified signals to efferent neurons which transfer them back to the cardiovascular system. Importantly, primary integration centers are wired to and receive information from secondary brain centers that control a wide variety of brain traits encoded in engrams including immune memory, stress regulating hormone release, pain, reward, emotions, and even motivated types of behavior. Finally, we explore the important possibility that brain effector neurons in the cardiovascular brain circuit network connect efferent signals to other peripheral organs including the immune system, the gut, the liver, and adipose tissue. The enormous recent progress vis-à-vis the cardiovascular brain circuit allows us to propose a novel neurobiology-centered cardiovascular disease hypothesis that we term the neuroimmune cardiovascular circuit hypothesis.

Keywords: brain, cardiovascular disease, heart, infarcts, neuron


Neural brain circuits can be broadly defined as hardwired polysynaptic axonal connections from peripheral tissues to the brain and back to peripheral tissues. However, defining neural circuits at the structural and functional levels turns out to be one of the most humbling but also rewarding challenges in current medical research. Delineating circuits that involve the brain requires molecular, structural and functional information of the interactions and connections of peripheral and central nervous system (PNS, CNS) neurons with - in principle - all peripheral tissues and indeed most organ cells. The human brain as the center of both interoception (perception from bodily signals) and exteroception (perception from the environment) is certainly the most complex tissue that nature has ever coined135. Indeed, if challenges can be overcome to make steps towards a better understanding of the brain, groundbreaking advances into human development and conceivably the pathogeneses of a large number of clinically important cardiovascular diseases (CVDs) may be within reach36. Defining control of physiological responses such as stress-related circuits have already uncovered aberrant interactions between the brain and peripheral organ dysfunction in diseases as diverse as cancer37, obesity24, metabolism and diabetes mellitus38, chronic lung diseases39, 40, inflammatory bowel diseases20, 41, autoimmune diseases42, psychiatric disorders43, 44, and neural control of infectious diseases3, 28. It is important to distinguish these types of circuits as hard-wired bona fide circuits from interactions mediated by soluble molecules including neurotransmitters and hormones produced by the immune, endocrine or the nervous systems, respectively45. Progress in deciphering these circuits has recently expanded at an accelerating pace46. How peripheral signals are being processed by distinct subgroups of sensory neurons in brain centers (also referred to as brain nuclei) such as the brainstem, the parabrachial nuclei, the hypothalamus and the insula have already provided mechanistic insight into multiple brain traits that had remained outside of any comprehension including the sickness syndrome47, fear and empathy48, jealousy, and autism and schizophrenia5, 8, 43, 44, 49. For few circuits, neuron subtypes that integrate these signals and connect to other neuron subtypes projecting back to the periphery have been identified in mice50. It is noteworthy that many circuits not only involve several constituents of the PNS and the CNS but that both interact with organ-specific relatively independently acting intrinsic nervous systems1, 26, 28, 5153.

Several recent lines of evidence point to a new - until recently elusive - circuit involving the arterial tree that we have termed the artery brain circuit (ABC)7, 8, 54. The ABC appears to interact with the much better established heart brain circuit (HBC) to form a large dynamic cardiovascular brain circuit (CBC)26, 27, 35, 55. In this review, we will discuss what is currently known about both the ABC and the HBC to form a global CBC network with its core constituents, i.e. the arterial tree and the heart. We will discuss that multiple types of CBC dysfunction directly affect a substantial number of CVD outcomes including myocardial infarcts (MIs), sudden death, acute and chronic heart failure, kidney dysfunction, stroke and dementia46. Finally, we will propose a novel neurobiology-centered CVD hypothesis: The neuroimmune cardiovascular circuit hypothesis.

THE ARTERY BRAIN CIRCUIT (ABC)

The conventional target of atherosclerosis research has been the inner layer of the arterial wall, i.e. the intima, because atherosclerotic plaques arise there5666. However, it is well established that the disease involves all three arterial wall layers with the media and adventitia layers also being affected9. Unlike mainstream research with its focus on the intima plaque, we decided early to focus on the adventitia which forms the outer connective tissue coat of arteries: We found that the majority of immune cells during disease progression in mice accumulated in the adventitia rather than in plaques54, 6774. Since it became apparent that the adventitia is the arterial wall layer where the immune system senses atherosclerotic plaques in the intima by forming aggregates of innate and adaptive immune cells, interest in the adventitia gained momentum. Later, delineation of artery tertiary lymphoid organs (ATLOs) in the adventitia of diseased but not healthy arteries in hyperlipidemic mice as well structured leukocyte aggregates that resembled those found in cancer and autoimmune diseases and other chronic inflammatory diseases raised further interest68, 69. Moreover, it became apparent that similar immune cell aggregates formed in atherosclerotic aortic aneurysms and coronary arteries in humans75 and that disruption of them in mice altered the progression of atherosclerosis69. The adventitia has been the subject of recent reviews and will therefore not be considered further here73, 74, 7679. Owing to space limitations, we will not cover extensive discussions of risk factors in this review as multiple recent reviews have been reported5660.

Arteries directly talk to the brain via polysynaptic axonal connections

The discovery of a structural ABC was made possible by several lines of evidence: In landmark observations made by the founder of modern anatomy, i.e. the Belgian anatomist Andreas Vesalius, who in 1543 described the striking macroanatomical proximity of the PNS and the arterial tree17. Research beginning in the late 20th century identified numerous interactions between the PNS, blood vessels and immune cells leading to two relatively independently studied areas: the area of the cardiovascular link13, 17, 31 and the area of neuroimmunology16, 2023, 80: imaging studies revealed that the PNS not only uses the adventitia as its main conduit to reach distant targets but that its axon endings directly gain access to the adventitia. Moreover, restructuring of the innervation of the adventitia was particularly pronounced in diseased segments of arteries: It involves newly formed axon networks54. Focusing on the abdominal aorta of aged ApoE-deficient mice, two components of the PNS were identified in the formation of neuroimmune cardiovascular interfaces in the adventitia: The sympathetic nervous system and the sensory nervous system but - in this segment - not the parasympathetic nervous system. Interestingly, this type and location of aorta adventitia innervation resembled that of normal lymph nodes81. However, segments of other arterial tree segments showed a different type of innervation pattern with the vagus nerve innervating the aortic arch26, 52, 53. In view of this apparent heterogeneity of artery innervation, we expect that future studies will identify further nervous system components in other artery segments depending on their respective functions and anatomical microdomains to regulate cardiovascular homeostasis.

The ABC Sensor

Two major PNS components were identified in the adventitia of diseased arteries of aged hyperlipidemic mice and human coronary arteries and abdominal aorta aneurysms: The sensory nervous system forms the ABC sensor and the sympathetic nervous system forms the ABC effector (Figures 1, 2). We interpret the restructuring events during atherosclerosis progression of both components of the ABC in the adventitia as a response of the PNS to the inflammatory environment afforded by the leukocytes that specifically infiltrate those adventitia segments that are burdened by plaques9, 69, 71. Multiple molecules that are involved in the bidirectional communication between the sensory nervous system are likely to be produced by leukocytes but also by the axon endings including calcitonin gene-related peptide and multiple nociceptors: our immunohistochemical analyses of the adventitia showed marked expression of transient receptor potential V112, 8284 at axon endings as well as markers of axon growth cones. The transient receptor potential V1 is a classical nociceptor that responds to a variety of exogenous stimuli including heat, the chili pepper ingredient capsaicin (though these are unlikely mediators of the ABC) but - with relevance to atherosclerosis and other CVDs - to inflammatory mediators that may be induced in activated immune cells during atherosclerosis progression (Figure 1). Using virus tracing studies we showed that the upper abdominal aorta segment, is innervated by neurons in dorsal root ganglia (DRG) from where the sensory nervous system enters the spinal cord to reach the parabrachial nucleus and the central amygdala54 (see for comparison below the distinct innervation patterns of the heart in the HBC in Figure 3). When taken together, this data established the sensory arm of the ABC.

Figure 1. The ABC sensor.

Figure 1.

We propose that the ABC is initiated in the adventitia of diseased artery segments establishing a tridirectional communication network between innate immune cells, the arterial wall, and the peripheral nervous system. Inflammatory mediators may activate nociceptive receptors, for example, TRPV1 (transient receptor potential vanilloid 1), ion channel receptor, for example, Nav1.8 (voltage-gated sodium channel 1.8), P2X3 (P2X purinoceptor 3), cytokine receptors and GPCRs (G protein-coupled receptors) at axon endings of sensory neurons resulting in action potentials that are transduced to dorsal root ganglia to enter the spinal cord to be conveyed to brain nuclei (see below).

Figure 2. The ABC effector.

Figure 2.

Sympathetic nervous system neurons are projected via the spinal cord to the periphery54. Sympathetic nerves of celiac ganglion neurons directly innervate the adventitia.

Figure 3. The HBC.

Figure 3.

The HBC is composed of a sensory arm through vagal and spinal afferents reaching the brain either via DRGs and the spinal cord or via the nodose ganglia; the motor arm contains sympathetic and parasympathetic efferents. The ICN communicates with the heart and the vagus and the sympathetic nervous system. The ICN is preferentially present in the atria (inset above the heart symbol). RA, right atrium; LA, left atrium; ICN, intrinsic cardiac nervous system.

The ABC Effector

The ABC effector of the sympathetic nervous system was identified using a combination of imaging and virus tracing experiments in hyperlipidemic mice54. There was a marked increase of adventitia tissue epinephrine levels (and calcitonin gene-related peptide levels for the ABC sensor) in areas that are burdened by plaques (Figure 2). This data may be interpreted as a biologically relevant interaction network between the arterial wall, the immune system and the nervous system to form neuroimmune cardiovascular interfaces85. Transcript analyses of these artery segments revealed the formation of multiple other neurotransmitters, cytokines, and growth factors as potential candidates for this interaction69, 71.

THE HEART BRAIN CIRCUIT (HBC)

A better understanding of the multi-directional communication system between the heart and the brain in health and disease will fill important gaps. This will not only improve our understanding of the physiology of heart innervation but also open the way to delineate the basis for a series of heart diseases including atrial fibrillation, life-threatening arrhythmias such as ventricular tachycardia and ventricular fibrillation with their severe forms leading to sudden death. There are several major components in the HBC86 (Figure 3): First, the sensory arm is composed of vagus afferents and spinal nerves from the heart; second, the CNS cardiovascular center in the brainstem integrates interoceptive signals and higher brain (examples are circadian clocks regulating heart rate) inputs that result in the generation of premotor outputs; third, extrinsic cardiac motor control through parasympathetic and sympathetic efferent nerves; and fourth, elaboration of local neural networks formed by neurons located within the heart which is also referred to as the intrinsic cardiac nervous system (ICN) (see below).

Sensing from the Heart

Signals from the heart and distinct artery segments such as the aortic arch are closely monitored by extrinsic sensory neurons in the vagus and spinal nerves, with their cell bodies located in the nodose ganglia or in DRGs (Figure 3); and for the arterial tree most likely all DRGs86, 87, 88. More work is needed to delineate the relation between sensory neurons in the DRG versus those in the nodose ganglion. As a major neuronal subtype, vagal sensory neurons actively monitor numerous vital heart-derived cues to maintain cardiovascular homeostasis26, 89, 90: pulsatile and tonic pressure changes are sensed at various locations such as atria, ventricles, and the aortic arch. In addition, vagal sensory neurons also monitor pathological changes like ischemia and inflammation, likely through secreted peptides and multiple signalling molecules89, 90. Vagal aortic afferents with diverse sensory modalities, response patterns, and activation thresholds have been described. At least two specialized atrial receptors have been characterized, both of which are fast conducting A-fibers but may differentially sense atrial systole and filling. The majority of vagal ventricular afferents are capsaicin-sensitive C-fibers that are also sensitive to alkaloids such as veratridine as well as bradykinin91. Together, the results have shown that cardiac sensation via the vagus nerve is complex, demonstrating the important role of the HBC in maintaining cardiovascular homeostasis. Such heterogeneity and complexity present a considerable challenge for studies to better understand the general coding principles for cardiac signals and the underlying molecular and cellular mechanisms for the majority of cardiovascular responses. However, considerable efforts are underway to delineate vagal cardiovascular afferents from anatomical, molecular, and functional angels. A major question is if and how sensory endings are morphologically specialized for the input signals. Using a collection of anterograde tracing approaches, a variety of specialized vagal sensory endings have been characterized52, 55, 92, 93. However, except for a clear link between aortic end-net endings and arterial blood pressure sensing52, signals sensed by these morphologically and genetically defined sensory axon endings largely remain unknown. Linking terminal morphologies with their electrophysiological properties and physiological roles for vagal cardiac afferents has been an extremely challenging task. However, delineation of them will undoubtedly provide new insights for better understanding the underlying sensory mechanisms implying major progress for future therapeutic interventions in diseases as varied as high blood pressure, arrhythmias and thrombus formation due to atrial fibrillation.

Activation of vagal cardiac afferents triggers physiological reflexes to regulate heart rhythm, blood pressure, cardiac output, respiration and renal functions to maintain cardiovascular homeostasis90. Sensations of blood pressure changes and oxygen/CO2 levels from the aorta via vagal aortic baroreceptor and chemoreceptor neurons regulate heart rate/blood pressure and respiratory patterns, respectively, similar to their carotid counterparts52, 53. The Bainbridge reflex, i.e. a tachycardia response to an increase of blood volume, is a regulatory reflex initiated by atrial stretch or distension detected by vagal sensory endings in heart atria. Like the baroreflex, the Bainbridge reflex is bi-directional in the sense that a decrease in venous return, such as during hemorrhage and hypotension, will decrease heart rate94. Thus understanding the Bainbridge reflex holds therapeutic potential to treat acute life-threatening conditions such as those experienced in the intensive care unit including shock or a low ventricular ejection fraction following a MI. Moreover, the Bezold-Jarisch reflex is an inhibitory cardiac reflex attributed to activation of vagal sensory endings on heart ventricles that causes severe bradycardia and hypotension95, 96. When taken together, despite their highly important roles in cardiovascular regulation in both healthy and disease conditions, our basic knowledge about most vagal cardiac afferents remains limited. However, recent advances in mouse genetics are rapidly improving our understanding of vagal cardiac afferents. Several genes have been proposed to be critically involved in the baroreflex9799, i.e. the mechanosensitive ion channels PIEZO1 and PIEZO2 fulfil most criteria of being the primary baroreceptor that directly senses blood pressure changes53. Knocking out both Piezo1 and Piezo2 from vagal sensory neurons in paired-like homeobox 2b-Cre mice resulted in an almost complete loss of the baroreflex. The same phenotype was also observed in mice with ablated Piezo2+ vagal sensory neurons. Moreover, optogenetic activation of Piezo2+ vagal sensory neurons induced pronounced bradycardia. Anatomically, Piezo2+ vagal afferents form macroscopic claws around the aorta, which are decorated with mechanosensory ‘end-net’ endings52. Surprisingly, it seems that ‘flower-spray’ endings are not involved in blood pressure sensing as ablating aortic ‘flower-spray’ endings did not impact the baroreflex. Therefore, it is apparent that aortic blood pressure is sensed by PIEZO1/PIEZO2 mechanosensitive ion channels through ‘end-net’ endings52, 53.

Similar to vagal afferents, cardiac DRG neurons also sense mechanical changes and a variety of neurochemicals, many of which are involved in cardiac pathological conditions such as myocardial ischemia and inflammation100. Stimulation of cardiac DRG afferents results in an increase of sympathetic tone and a decrease of parasympathetic tone. For example, electrical or chemical stimulation of cardiac sympathetic afferents blunted the arterial baroreflex101. Elevated cardiac DRG afferent activity has been described in CVDs including chronic heart failure and hypertension, and their roles have been extensively studied using various animal models.

Cardiovascular Control from the Brain

The brain directly communicates with the heart through two major routes: the parasympathetic pathway involving vagal motor neurons in the brainstem and postganglionic neurons in the heart; and the sympathetic pathway involving preganglionic neurons86, 102. In general, parasympathetic outputs mediate negative chronotropic effects through cholinergic signalling while sympathetic outputs positively impact both chronotropy and inotropy via adrenergic signalling. Here we will focus on some recent results on circuit mapping.

Vagal motor neurons are primarily located in two brainstem nuclei: the dorsal motor nucleus of the vagus and the nucleus ambiguus. However, the molecular mechanisms for axon targeting for vagal cardiac motor neurons remain to be elucidated. Single cell studies using scRNAseq approaches have identified 8 dorsal motor nuclei of the vagus neuron subtypes, with distinct molecular markers and spatial distribution patterns103.

Sympathetic postganglionic neurons densely innervate the sinoatrial and atrioventricular nodes and directly modulate pacemaker activity via norepinephrine release that acts primarily through the β1 adrenergic receptor (ADR) and downstream cAMP signalling104, while other ADR types also contribute105. In addition to norepinephrine, sympathetic neurons also co-release neuropeptide Y which binds to the neuropeptide Y2 receptor expressed on the presynaptic terminals of cholinergic neurons to inhibit parasympathetic inputs to pacemakers104. Sympathetic nerves also extensively innervate atrial and ventricular cardiomyocytes and modulate cardiac contractility. Under disease conditions such as MI, sympathetic nerves undergo extensive remodelling, and such abnormal sympathetic activity may lead to arrhythmia and sudden cardiac death106.

The Intrinsic Cardiac Nervous System (ICN)

Extensive local neural networks in the heart suggests that neurocardiac control is far more complex than previously recognized. Neurons of the heart, so-called intrinsic cardiac neurons with cell bodies residing in dispersed clusters called ganglionic plexuses, form the ICN30, 86, 104, 107. This local neural network interacts with extrinsic afferent and efferent fibers to coordinate the heart-to-brain axis. Moreover, the ICN is able to act independently of extrinsic cardiac neurons as local actors. Both anatomical and electrophysiological evidence suggests that, like the enteric nervous system, a single ICN cluster might be composed of sensory, motor, and local circuit neurons that form a microcircuit to sense and process surrounding cardiac changes and control local cardiac functions. Moreover, ICN clusters among different cardiac ganglionic plexuses communicate with each other through interganglionic neurons to coordinate cardiac functions across the entire heart (Figure 3 inset).

In mammals, the number of ICNs is broadly correlated with the size of the heart (number of ICNs per heart: ~1,000 in mouse, ~6,500 in rat, ~12,000 in pig, and 43,000 – 94,000 in human)30. ICN clusters are discretely located on the dorsal atrial surface in small animals, but more profusely distributed across broader atrial regions and even extending to ventricles in larger animals including rabbit and human30, 108, 109. The majority of ICNs reside on the supra-ventricular epicardial surface30, 86, 104. Recent development in mouse genetics and adeno-associated virus-guided anatomical tracing has provided important tools. For example, pacemakers in the sinoatrial and atrioventricular nodes represent an important downstream target of ICNs. Electrical stimulation of cardiac plexuses resulted in a significant drop in the heart rate. On the other hand, surgical ablation of certain ICN ganglia blocks the bradycardia induced by vagus nerve stimulation. This ICN-sinoatrial node projection has been demonstrated in mice using a combination of genetically engineered animals (choline acetyl transferase-Cre mice) and specifically designed adeno-associated viruses that selectively target peripheral neurons110, and high-resolution volumetric light-sheet imaging of tissue-cleared hearts111. Meanwhile, activating specifically designed adeno-associated viruses targeting cholinergic ICNs located at the posterior region of the heart using optogenetics causes significant bradycardia. Together, this provides important evidence for the ICN-sinoatrial node connections from both anatomical and functional aspects at cellular resolution. In addition to the sinoatrial and artrioventricular nodes, ICNs likely cover a much larger heart area. Extensive cholinergic innervation of atrial appendages, interatrial septum, and ventricles has been characterized in various animal species108, 112. However, complete mapping of ICN projections on the heart remains to be an important challenge of future studies. Multiple ICN neuron subtypes with distinct morphologies, response properties, and physiological roles have been characterized. Immunohistochemical studies have revealed that a variety of neuromodulators and neurotransmitters are present in the ICNs30, 104, 113115. Although it has been well documented that different neurochemicals can be co-released, such as norepinephrine and neuropeptide Y in sympathetic neurons116, it is unclear whether neuropeptide Y or cocaine and amphetamine-regulated transcript-derived protein are also co-released with acetylcholine, and if so, how they differentially regulate heart functions. In a separate study using laser capture and microdissection, 154 genes were identified in 151 captured single ICNs from spatially distinct ganglionic plexuses117. Therefore, a detailed molecular architecture of the ICN network has not been established yet.

ICNs have been classified into different subtypes based on their distinct electrophysiological properties such as firing patterns113, 118120. Interestingly, whether some isolated ICNs have spontaneous rhythmic activities resembling pacemaker cells remains controversial. Electrical or pharmacological stimulation of ICNs resulted in multifaceted, frequency and location-dependent cardiac hemodynamic changes, suggesting that cardiac regulation by the ICN is under precise spatial and temporal control104. For example, stimulating selective ICN ganglia may elicit bradycardia, tachycardia, or more complicated changes in heart rhythms121. Local stimulation of ICNs within a ganglionic plexus may also modulate remote atrial and ventricular tissues, likely mediated by long-range projecting motor neurons or through neural circuits among multiple ganglionic plexuses. Although traditionally been thought as parasympathetic ganglia, it was estimated through electrophysiology that only ~40% of ICN receive direct parasympathetic inputs from the vagus nerve122. On the other hand, in vivo electrophysiological recordings of ICN in anesthetized dogs demonstrated that some ICNs may receive direct or indirect sympathetic inputs from the stellate ganglion109, 123.

A variety of CVDs are associated with malfunction of the ICNs, including atrial and ventricular fibrillation, myocardial ischemia, and heart failure86, 124. For example, abnormal activation of intrinsic cardiac ganglia around the pulmonary veins resulted in atrial arrhythmia, and ICN ablation is a viable option for intervention to treat atrial fibrillation in the clinic. Although the initiation of atrial fibrillation in humans is thought to be associated with an imbalance between sympathetic and parasympathetic tones, a subset of atrial fibrillation events can be triggered by abnormal ICN activity without extrinsic cardiac inputs125. Nevertheless, the underlying molecular and cellular mechanisms between aberrant ICN activities and atrial fibrillation remain an important area to develop therapeutic strategies in the area of cardiac arrhythmias. However, since ICN ganglia plexuses are heavily interconnected with each other, and various ICN subtypes may differentially regulate distinct heart functions, whether surgical ablation of predicted ICN ganglia plexuses in severe bradycardia remains to be determined126. Furthermore, mechanical or chemical disruption of cholinergic ICNs also increases the likelihood of ventricular arrhythmia127, which is a leading cause of sudden cardiac death under a variety of disease conditions. However, as electrical or chemical stimulation of selective ICNs triggers ventricular arrhythmias128 reveals that ICN activities need to be precisely regulated to maintain cardiac homeostasis. Other data further showed afferent and efferent remodelling of ICNs after MI, suggesting a functional role of the ICN in MI and heart failure. Thus, a better understanding of the biology of ICNs is likely to provide new therapeutic targets for treating CVDs including life-threatening arrhythmias under a variety of disease conditions.

THE IMMUNE SYSTEM IN ARTERY AND HEART HOMEOSTASIS AND DISEASE

The immune system emerged as a critical regulator of heart and artery physiology and CVDs. Studies combining flow cytometry, intravital microscopy and mouse genetic reporter models revealed that the heart harbours a broad repertoire of tissue-specific immune cells. The latter include macrophages, mast cells, monocytes, neutrophils, innate lymphoid cells (ILCs), dendritic cells, B cells and T cells. Cardiac macrophages are abundant, ontogenically diverse and heterogeneous. Four main cardiac macrophage subsets have been identified and each one occupies specific anatomical locations. For example, macrophages and dendritic cells are found in the aortic valves and coronary arteries; the atrioventricular node contains high numbers of macrophages; the pericardial fluid harbours macrophages, mast cells and B cells; and the pericardial adipose tissue contains a diverse set of leukocytes. Importantly, recent studies have shown that cardiac resident immune cells actively contribute to cardiac development and steady-state physiology throughout life, e.g., electrical conduction and cardiac rhythm129. In the context of CVDs, innate immune cells have been described to initially drive a pro-inflammatory response to overcome tissue damage and to promote wound healing at later stages of inflammation. Recruited and tissue-resident innate and adaptive immune cells have been shown to be involved in inflammation, revascularization, cardiomyocyte dedifferentiation and fibrotic scar formation in the injured cardiac tissue. The heterogeneity and role of cardiac immune cells in homeostasis and heart pathology, including MI, conduction disorders, myocarditis and endocarditis, have been extensively reviewed elsewhere129, 130.

The arterial wall also contains resident innate immune cells, including macrophages and dendritic cells, but their role in artery physiology only recently started to be acknowledged. In contrast, monocytes, macrophages and neutrophils, are well known for their roles in the initiation, progression, and destabilization of atherosclerotic lesions131. Over the past three decades our understanding of atherosclerosis has switched from a simple cholesterol-storage disease to a lipid-driven inflammatory disease. Notably, it is now well established that the chronic inflammatory disease of arteries depends on inflammatory leukocyte supply from the bone marrow and extramedullary sites. In addition to monocytes, macrophages and neutrophils, recent evidence suggests a potential involvement of previously underappreciated cell types in atherosclerosis, namely, dendritic cells and platelets132. Similarly, diverse adaptive immune cells, such as T cell subsets (CD8+ T cells and CD4+ regulatory T cells) have been shown to promote or attenuate arterial disease in rodent models133. Whether atherosclerosis and other CVDs is ordinary autoinflammatory disease as previously thought or whether autoimmune B cells and autoimmune T cells participate in the progression of atherosclerosis is currently emerging as an important new research focus134, 135. In murine atherosclerosis models and human diseased tissues, recent evidence is pointing to a bona fide autoimmune component for B cells136 and more recently autoimmune T cells133, 137, 138. This data has important implications for understanding the poorly understood relation between bona fide autoimmune diseases such as systemic lupus erythematosus or rheumatoid arthritis and atherosclerosis known to exacerbate each other139142. There is recent progress in the area of the role and a detailed description of the contribution of different immune cell subsets to atherosclerotic lesion progression which can be found elsewhere131, 132.

Despite the enormous progress towards the characterisation of cardiovascular immune cells, the cellular and molecular circuitries that underlie their function remain unclear. Given that cardiovascular immune cells are strategically located in close proximity to non-immune cells, such as cardiomyocytes, fibroblasts, endothelial cells, coronary microvessels, lymphatic vessels and nerves, it is very likely that interactions between immune and non-immune populations contribute to cardiovascular health and disease54. We will focus below on the fast-evolving field of neuroimmune interactions and their potential involvement in cardiovascular physiology and pathology.

Neuroimmune Cell Interactions in Peripheral Organs

Recent technological advances have provided new insights on cellular and molecular mechanisms that underly the communication between the nervous and the immune systems. Over the last decade, numerous studies have revealed that immune cells and nerve terminals colocalize and functionally interact at discrete anatomical locations in peripheral organs, forming neuroimmune cell units that orchestrate host defence, tissue homeostasis and organismal physiology143. These bidirectional neuroimmune interactions have been described across several organs, including the bone marrow, spleen, skin, lung, intestine and adipose tissue1, 2, 144 and – more recently in the adventitia of aged atherosclerotic mice54. In addition, such neuroimmune circuits are key orchestrators of multiple physiological processes, including haematopoiesis, organogenesis, host defence, inflammation, tissue repair, metabolism and thermogenesis1, 2, 143, 144. Different branches of the PNS, including the sensory, sympathetic, parasympathetic, and enteric nervous systems have been shown to form neuroimmune hubs, mostly with innate immune cells. While peripheral neuroimmune cell interactions have been extensively reviewed elsewhere1, 2, 143, 144, here our goal is to provide examples of neuroimmune interactions by macrophages and ILCs, which highlight the potential impact of neuroimmune regulation of cardiovascular health and disease.

Tissue-resident macrophages in the intestine and adipose tissue have been described to function as important hubs that integrate neuronal-derived cues. Under steady state conditions, enteric neurons maintain muscularis macrophages through secretion of colony-stimulating factor-1, which is required for macrophage development145. Reciprocally, muscularis macrophages directly regulate neuronal function via secretion of bone morphogenetic protein-2, which activates bone morphogenetic protein receptor-expressing enteric neurons145. This bidirectional neuronal-macrophage crosstalk regulates the peristaltic activity of the colon. Additionally, commensal microbe-derived signals can modulate bone morphogenetic protein 2 and colony stimulating factor 1 expression by macrophages and enteric neurons, respectively. This suggests that an intricate microbiota-driven neuronal-macrophage axis controls gastrointestinal motility, contributing to tissue homeostasis under steady-state conditions145. In the context of enteric bacterial infection, activation of extrinsic sympathetic ganglia enhances the tissue-protective profile of muscularis macrophages, via norepinephrine signaling in ADR2-expressing macrophages146. Interestingly, muscularis macrophages have also been shown to play a role in the maintenance of intrinsic enteric-associated neurons during enteric infection147. Specifically, ADR signalling in muscularis macrophages was shown to induce neuronal protection through an arginase 1-polyamine axis147.

In adipose tissue, a discrete population of macrophages tightly associates with sympathetic fibres. These neuron-associated macrophages take up and catabolize norepinephrine through expression of the norepinephrine transporter and the norepinephrine-degrading enzyme monoamine oxidase A, respectively148. During aging, a similar macrophage-mediated norepinephrine-degrading activity was reported in the adipose tissue149. Tissue-resident macrophages were also found to regulate sympathetic innervation of the brown adipose tissue, thus indirectly controlling local norepinephrine bioavailability, thermogenesis, and obesity150.

Macrophages are among the most abundant, well studied and functionally relevant - though endowed with diverse and sometimes opposite functions - immune subsets in heart physiology and in atherosclerotic plaques, regulating plaque growth. Despite intensive research, the potential of neuroimmune crosstalk in cardiac homeostasis and in the pathogenesis of CVDs remains largely unexplored. In the context of atherosclerosis, it has been shown that a neuroimmune guidance cue, i.e. netrin-1, and its receptor, are expressed by macrophages of human and mouse atherosclerotic plaques151. Selective deletion of netrin-1 in bone marrow-derived cells reduced atherosclerotic plaque size and complexity in mice deficient in the receptor for low-density lipoprotein, and promoted macrophage egress from plaques, possibly by inactivating the migration of macrophages toward the chemokines CC-chemokine ligand 12 and CC-chemokine ligand 19, which drive macrophage egress from plaques151. Still, evidence for direct and functional neuronal-macrophage crosstalk and its implications for heart and artery physiology and CVD is essentially lacking.

Akin to macrophages, ILCs are innate immune effectors that actively engage in neuronal crosstalk in peripheral tissues. At mucosal barriers, such as the intestine and lung, ILCs reside in close proximity to neuronal cells. In the intestine, a neuroglia-ILC3-epithelial axis promotes intestinal tissue repair upon inflammatory and infection insults152. Enteric glial cells sense microbial and host-derived alarmin cues and produce neurotrophic factors that act on tyrosine kinase receptor-expressing ILC3s, promoting the production of tissue-protective interleukin-22152. Enteric and pulmonary ILC2s have been also shown to integrate and respond to neuronal cues derived from cholinergic, sympathetic and sensory nerve fibers. Cholinergic neurons control ILC2-mediated responses to allergens or helminthic infections via production of neuromedin U153155. Neuromedin U signals through neuromedin receptor 1 expressed in ILC2s leads to a rapid and potent production of the type 2 inflammatory cytokines, interleukin-5 and interleukin-13, and of the tissue-protective amphiregulin, conferring immediate tissue protection to helminthic infection153155. Pulmonary and intestinal ILC2s also express the ADR2 and colocalize with adrenergic neurons in the intestine156. Abrogation of ADR2-mediated signalling results in increased ILC2 responses, type 2 inflammation, and lower helminth infection burden, effects that are reversed by ADR agonist treatment156. In the lung, vasoactive intestinal peptide released by nodose afferent nociceptor sensory fibres potentiates airway inflammation by stimulating ILC2s to produce interleukin-5, which can act directly on nociceptors, further increasing vasoactive intestinal peptide expression and generating a type 2 inflammatory signalling loop that potentiates allergic inflammation157. Interestingly, ILC2s have recently been shown to promote heart tissue repair, to alleviate cardiac fibrosis upon acute cardiac injury and to improve the recovery of heart function following MI158, 159. Nevertheless, whether neurons and ILCs co-operate to orchestrate cardiovascular physiology in health and disease remains unknown.

Long-Distance Neuroimmune Interactions

Recent studies have started to unravel how neuroimmune interactions regulate health and disease via brain-body axes. These studies are providing new insight into the brain regions involved in these complex regulatory networks and are revealing the mechanistic pathways through which neuroimmune circuits operate at an organismal level. It has recently been shown that light-entrained and brain-tuned circadian circuits regulate enteric ILC3s, intestinal homeostasis, gut defence and host lipid metabolism160. Surgical- and tissue-specific genetic approaches identified the suprachiasmatic nuclei in the hypothalamus as a key regulator of enteric ILC3 homeostasis, intestinal physiology and health160. In another study, neuro-mesenchymal units were shown to control ILC2s and obesity via a brain-adipose circuit161. Combined retrograde tracing and functional chemogenetic manipulations identified a sympathetic aorticorenal circuit that modulates ILC2s in gonadal fat and connects to higher-order brain areas, including the paraventricular hypothalamic nucleus in the hypothalamus161. Thus, neuro-mesenchymal units translate cues from long-distance neuronal circuitry into adipose-resident ILC2s function, shaping host metabolism and obesity161. In the context of adaptive immune responses, a brain-spleen circuit was shown to directly impact the formation of immunoglobulin-producing plasma cells via a paraventricular hypothalamic nucleus / central amygdala-splenic nerve -B cell axis50. Stress-responsive neurons in the central amygdala and the paraventricular hypothalamic nucleus that express corticotrophin-releasing hormone were shown to be connected to the splenic nerve and pharmacogenetic activation of these neurons increased plasma cell formation after immunization50. Interestingly, in the context of psychological stress, distinct brain regions were shown to differentially and rapidly shape leukocyte distribution and function across peripheral organs, impacting on disease susceptibility162. Motor circuits induced rapid neutrophil egress from the bone marrow to peripheral organs via skeletal-muscle-derived neutrophil-attracting chemokines, while corticotrophin-releasing hormone neurons in the paraventricular hypothalamic nucleus promoted monocyte and lymphocyte mobilization from blood and secondary lymphoid organs to the bone marrow through direct, cell-intrinsic glucocorticoid receptor signalling, thus protecting against autoimmunity, but impairing immunity to viral infections162. Finally, intestinal inflammation activates neurons in a brain area called the insular cortex or the insula. Artificial reactivation of these ‘immune-imprinted’ neurons is sufficient to generate organ-specific recall of inflammatory responses that resemble the initial inflammatory episode163. Altogether, these and other studies (reviewed in25) are raising increased interest in mapping the brain regions and the communicating pathways involved in brain-peripheral immune system communication. Nevertheless, whether long-distance neuroimmune circuits are homeostatic cardiovascular players largely remains elusive.

Lifestyle Factors

Long-distance neuroimmune circuits allow for the integration of external challenges (exteroception) into internal body homeostasis (interoception) as introduced above. How lifestyle factors, such as sleep, psychological stress, diet and exercise, are integrated in cardiovascular physiology, possibly by neuroimmune circuits, also largely remains unexplored. Both epidemiological evidence and data from animal models indicate that stress and high-fat/high-cholesterol diet aggravate chronic inflammatory diseases, while regular exercise and healthy sleeping habits help to prevent CVDs164. It is becoming increasingly clear that lifestyle-derived factors shape haematopoiesis, leukocyte trafficking, and innate immunity165167. In agreement with these views, several studies unravel the pathways through which sleep, stress, diet and exercise modulate haematopoiesis and promote inflammatory leukocyte supply to the atherosclerotic lesion, accelerating disease progression168170. Interestingly, some of these studies have revealed neuroimmune pathways as the underlying mechanism linking lifestyle factors to CVD progression. As an example, it has been shown that sleep regulates haematopoiesis and protects against atherosclerosis in mice via a neuroimmune axis. Mice subjected to sleep fragmentation display reduced levels of hypocretin in the lateral hypothalamus. This stimulatory and wake-promoting neuropeptide regulates myelopoiesis and protects against atherosclerosis by limiting the production of macrophage colony-stimulating factor-1 by hypocretin-receptor-expressing pre-neutrophils in the bone marrow171173. In humans, autoimmune destruction of hypocretin causes narcolepsy and low plasma levels of hypocretin are associated with increased risk of MI and heart failure. Uncovering how lifestyle factors and cardiovascular neuroimmune circuits intersect to shape cardiovascular health, disease onset and progression is certainly amongst the most challenging tasks ahead.

Inputs from the ABC and HBC Merge in Distinct Brain Nuclei

Brain nuclei are clusters of neurons at a specific location in the brain containing a variety of neuron subtypes which receive afferent signals or target peripheral organs. When we compared larger brain areas involving the ABC or the HBC, we noticed that both subcircuits shared several brain nuclei (Figure 5)26, 36, 54, 55, 174182. This observation raised an important new question: Are identical neuron subtypes shared by ABC and HBC afferents and/or efferents within these nuclei ? If this were to be the case, it would indicate that specific neuron subtypes may be concomitantly activated by the ABC and the HBC depending on physiological and/or pathophysiological inputs implying direct ABC-HBC interactions at the level of single brain neurons. To answer this important question, a series of experimental approaches using new mouse models and an extensive set of cutting-edge methods need to be employed. These studies will define the connectivity networks of the ABC and the HBC and identify the nature of their interactions and places within the global CBC connectome. Understanding the CBC connectivity map in the brain and the periphery may contribute to mechanistic insights into the atherosclerotic brain before life-threatening types of brain injury such as strokes ensue.

Figure 5. The ABC and the HBC share large functionally diverse brain areas but whether identical neuron subtypes are shared remains unknwon.

Figure 5.

Major multifunctional and markedly heterogeneously brain areas are shared by the ABC and the HBC. However, whether neuron subtypes connect to arteries and the heart remains to be defined. Here we list broad functions that have been assigned to these brain nuclei with the potential to regulate CVDs: Insula (emotions)5, 6, 36, 183, 184, Amygdala (memory)175, 185, Hypothalamus (stress)186, Rostral Ventrolateral Medulla179, 187 (stress), Nucleus Tractus Solitarius (body weight)26, 188, and Dorsal Motor Nucleus of the Vagus (metabolism)55, 188. Many other impacts and connections of these nuclei are not depicted for ease of reading.

ARTERIES AND THE HEART FORM A GLOBAL CBC NETWORK AND ARE ORGANIZED IN ENGRAMS

Recent progress to define a structural ABC7, 8, 54 together with the better understood HBC26, 55, 189, 190 and rapid advances on the role of the immune system to direct both the ABC and the HBC1, 2, 9, 69, 71, 133, 143, 191 support the presence of a global CBC network in which multiple cardiovascular subcircuits interact (Figures 17). When the known components of this network are considered from the point of view of neuroimmune circuit physiology and pathophysiology, several points deserve attention: The interaction between arteries and the heart to form a CBC is only beginning to be understood as the two have largely been studied by relatively independent areas in the past. While atherosclerosis research has focused on the atherosclerotic plaque in the intima5658, 60, 192 and on molecular mechanisms of atherosclerosis risk factors including hyperlipidemia and lipid transport, hypertension, diabetes mellitus, obesity, lifestyle, research of the heart has been dedicated to physiological responses, MI healing and treatments of arrhythmias and heart failure. Now, time has come to bring artery and heart innervation together and begin to appreciate the role of the brain to power the CBC network and its subcircuits1, 5, 25, 26, 36, 55, 191, 193. Broadly, the CBC can be functionally divided into CBC sensors (Figure 6) and CBC effectors (Figure 7). It should be noted that each of the participants of the CBC consist of multiple genetically, functionally and territorialized distinct subcircuits to provide well organized neuronal control nodes depending of specific requirements of the various artery segments and heart compartments. What may be the connectivity structure of these subcircuits ? Each subcircuit may be defined what has been termed engrams (the term engram was originally coined for bona fide memory neuronal networks) which can be described as assemblies of neurons that share inputs from or outputs to peripheral stimuli194. Engrams are known to exceed the borders of a single brain nucleus by forming a collection of neurons stretching across several nuclei in a network-like pattern194. The identification of engrams in the CBC has not yet been accomplished directly but their existence is likely given the involvement of the immune system in forming engrams in other disease conditions and being involved in CVD progression: Thus, brain engrams have been shown to exist for physiological but also for pathophysiological responses involving the immune system58, 43, 44, 46, 49, 54, 163, 193, 195 forming inflammation-related engrams that are defined as the ability of the brain to remember past tissue injury of any given peripheral organ6, 163, 196. Therefore, brain engrams can also be defined as regulatory units of neurons that store information of past physiological organ-to-brain connections or organ destruction. Of note, engrams endow important networking tasks to regulate longterm brain-organ responses. Furthermore, new engrams may form throughout life to adjust to the needs of peripheral organ physiological responses that are controlled by the brain. For all these reasons, we view the identification of CBC engrams as one of the major prerequisites to study the impact of activating or inactivating distinct neurons to define their roles in driving or preventing CVDs. Understanding potential cardiovascular engrams in the framework of the neuroimmune cardiovascular circuit hypothesis is expected to open unprecedented opportunities to test the role of distinct neuron subtypes to affect CVD progression. To delineate these engrams a powerful tool box is available in mice facilitating translational and clinical trials.

Figure 7. Brain to organ connections form a CBC effector and yield engram-derived cues back to the cardiovascular system.

Figure 7.

Retrograde virus tracing from the adventitia of the abdominal aorta segment revealed direct polysynaptic efferent connections of autonomic brain nuclei via the brainstem and the spinal cord to the celiac ganglion54, while similar studies of heart innervation showed vagus efferent neurons in the brain stem innervating the aortic arch and the heart including the atria (location of the ICN) and ventricles106, 114. In addition, it is known that spinal cord sympathetic nervous system innervation involves the bone marrow, the adrenal gland, the spleen and other internal organs. These brain-to-organ efferents contain information from cardiovascular injury or other types of past tissue injuries what can be defined as engrams6, 36, 163, 196, 197.

Figure 6. Organ to brain connections form a CBC sensor and program engrams.

Figure 6.

Focusing on the upper abdominal aorta, virus tracing has shown that the adventitia is directly innervated by neurons of DRG segments T6–1254, while sensory neurons in the nodose ganglia of the vagus nerve and different DRGs innervate the heart (Figure 3)87, 88, 26, 52, 55, 92, 93. Both routes of cardiovascular innervation gain access to the brainstem to be projected to higher brain areas along the pain pathway54. The chronic flow of organ-to-brain signals may form sensory engrams of the cardiovascular system.

Unprecedented Opportunities to Treat CVDs May Emerge from Understanding the Large CBC Network

As the CBC has only recently emerged as a hardwired polysynaptic nervous system axonal network, we can only imagine what is going on behind (not yet supported by data) closed doors. Progress in this area of neurobiology, however, is needed to develop preventive and therapeutic approaches to understand root causes of CVDs. Several principle aspects may come from preclinical models: Direct interference into the impact of neuron subsets in the brain to directly determine the longterm effect of the activity state of these neuron subtypes on a variety of CVD outcomes including atherosclerosis, mechanisms of arrhythmias, myocardial healing, ischemia perfusion injury are needed. Moreover, the PNS afferents and efferents within the CBC require analyses using electrophysiological approaches to identify nerve territories to apply localized surgical approaches as reported recently in a mouse model54. Within the brain compartment of the CBC, defining primary and secondary integration centers including the brain endocrine system will yield further mechanistic insights. Mapping distinct neuroimmune cardiovascular interfaces by scRNAseq approaches such as those already applied to the adventitia or the ICN may lead to the identification of molecular cues such as neurotransmitters, inflammatory mediators or receptors expressed on neuronal cells or nonneuronal cells that participate in the CBC. Such advances will help to identify their functions in the cardiovascular system and guide translational studies in human brains and the human PNS. Therefore, we expect that the identification of new tangible experimental approaches and of new CBC-related biomarkers in the circulation from a combination of basic research and clinical studies in the coming years.

A State-of-the-Art Toolbox is Required to Uncover the Structure and Function of the Global CBC Network: Warm up for a Marathon

While there is considerable progress in defining neuronal circuits in experimental mice, studies of both the ABC and the HBC to form a global CBC is in its infant stages. While the toolbox (Figure 8) to delineate the CBC further will allow to answer urgent questions as related to the identification of neuron subtypes and address the impacts of neuron subtypes on CVD progression, multiple additional methods and approaches have to be employed including the generation of new mouse models that allow to assay both atherosclerosis progression and heart function in both short term and long term experiments. These mouse models may combine distinct heart-related transgenic and tissue-specific mouse models52, 53, 55 on a hyperlipidemic genetic background54, 198. In particular, translational studies including clinical trials as they relate to the CBC are demanding tasks as the former are at present largely limited to morphological approaches using a variety of descriptive imaging tools. Yet, the combination of functional magnetic resonance imaging and machine learning algorithms provide new approaches to identify neuronal cells forming engrams197. However translational studies will come into further focus if work in mice will yield testable hypotheses in CBC research. This work is predicted to come from postmortem studies of human neuronal tissues using single nuclear RNAseq analyses199, 200, conventional immunohistochemical analyses and multiple brain imaging approaches201, 202. We highlight here six major core methods that are all required to make progress to investigate the CBC (Figure 8).

Figure 8. Toolbox to investigate the CBC.

Figure 8.

Progress in understanding the CBC requires state-of-the-art tools that exploit neurobiology approaches including tissue clearing203, multiomics133, 204, optogenetics184, 205, 206, chemogenetics207, 208, electrophysiology54, 55, 209, and multiple tracing methods54, 55, 161, 163. We refer to excellent recent reviews covering each of the methods here for the interested reader.

THE NEUROIMMUNE CARDIOVASCULAR CIRCUIT HYPOTHESIS

Multiple hypotheses have been suggested in the past to address the multifactorial nature of the pathogenesis of atherosclerosis as the major pathology underlying CVDs, including the lipid storage/retention hypothesis, the response to injury hypothesis, the oxidized low density lipoprotein hypothesis, the immune and inflammation hypothesis among others yet none of them has provided therapeutic options in the clinic56, 58, 61, 129, 131, 210213. As reviewed above, recent research into the pathogenesis of atherosclerotic coronary artery disease and other segments of the arterial tree together with a better understanding of the innervation of the heart suggests the existence of an ABC and an HBC1, 6, 25, 26, 36, 46, 5255, 163, 184, 214. Moreover, the ABC and the HBC form a global CBC network regulating both physiological and pathophysiological responses of the cardiovascular system. The understanding of the workings of the CBC is still in an infant stage requiring application of state-of-the art toolboxes to progress (Figure 8). Here, we propose a new comprehensive hypothesis of the physiology and pathophysiology of the cardiovascular system with the nervous system as its core element that we term the Neuroimmune Cardiovascular Circuit Hypothesis: In this hypothesis, the brain is viewed as the principal hub in which both genes and interoceptive and exteroceptive cues including risk factors are received, processed and integrated195. Furthermore, it integrates previous hypotheses related to inflammation and the immune system.

CONCLUSIONS

Recent progress in both basic and translational research into the anatomy and function of artery and heart physiology has established the contours of a CBC consisting of two major essential components: the ABC and the HBC. Within the CBC, the brain acts as a forceful instructor, master switch and control hub to regulate physiological responses. The complexity of the nervous system combined with the multilayered cardiovascular and immune systems makes the CBC highly vulnerable, however, to trigger CVDs when homeostatic regulation goes awry. A further disease susceptibility relates to the brain as an integration hub not only for interoceptive inputs from bodily signals but also for exteroceptive cues from the environment. In this regard we propose to view exteroception in the context of the common CVD risk factors such as unhealthy diets, sedentary lifestyles, and other harmful participants that manifest themselves as high blood pressure, type II diabetes mellitus, obesity and hyperlipidemia. When the wide variability of the genetic makeup of each individual human is taken into consideration with the above, a new holistic hypothesis regarding the mechanisms that control CVDs may be considered. We propose to tentatively name this hypothesis the neuroimmune cardiovascular circuit hypothesis (Figure 9).

Figure 9. The Neuroimmune Cardiovascular Circuit Hypothesis.

Figure 9.

The neuroimmune cardiovascular circuit hypothesis emphasizes the central role of the brain in affecting CVD progression. It incorporates several components: The interoceptive impacts of arteries and the heart on the CBC in addition to the genetic background and lifestyle exteroceptive impacts on each individual human. Depending on the composition of these inputs and actions of - at times opposite - factors, cardiovascular homeostasis may be either maintained or disrupted to accelerate CVD progression.

Figure 4. Neuroimmune interactions regulate CVD progression via long-distance brain-body axes.

Figure 4.

Long-distance neuroimmune circuits allow for the integration of exteroceptive and interoceptive cues in health and disease. Bidirectional connections between immune cells and the nervous system empowers neuroimmune axes with the unique capacity to efficiently perceive, integrate and respond to the ever changing external and internal environmental perturbations. We hypothesise that neuroimmune circuits operate in the heart and arteries to orchestrate cardiovascular physiology, health and disease.

Acknowledgements:

We apologize to our colleagues whose work we were unable to cite because of space constraints.

Sources of Funding:

This work has been supported by Deutsche Forschungsgemeinschaft (DFG): MO 3054/1-1, SFB1123-Z1, and Corona Foundation grant to S.K.M.; DFG YI 133/3-5, to C.Y.; DFG HA 1083/15-5 and European Research Area Network on Cardiovascular Diseases (ERA-CVD, PLAQUEFIGHT) 01KL1808 to A.J.R.H.; DFG Cluster of Excellence SyNergy (EXC 2145 SyNergy 390857198) to C.W.; and Easemedcontrol R & D Gmbh & Co KG to S.K.M, C.Y. and A.J.R.H.. National Institutes of Health (NIH) R01HL150449, R01AT012041, and DP2HL151354 to R.B.C. Fundação para a Ciência e Tecnologia (FCT) to C.G.-S.. European Research Council (ERC), Paul G. Allen Frontiers Group, Chan Zuckerberg Initiative, La Caixa, and FCT to H.V.-F..

Nonstandard Abbreviations and Acronyms:

ABC

artery-brain circuit

CBC

cardiovascular brain circuit

CNS

Central Nervous System

DRG

dorsal root ganglion

HBC

heart-brain circuit

ICN

intrinsic cardiac nervous system

ILC

innate lymphocyte cell

MI

myocardial infarct

NA

nucleus ambiguus

PNS

peripheral nervous system

scRNAseq

Single Cell RNA sequencing, method to identify the transcriptome of single cells

Footnotes

Disclosures

There is no conflict of interest.

References

  • 1.Klein Wolterink RGJ, Wu GS, Chiu IM and Veiga-Fernandes H. Neuroimmune Interactions in Peripheral Organs. Annual review of neuroscience. 2022;45:339–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Huh JR and Veiga-Fernandes H. Neuroimmune circuits in inter-organ communication. Nature reviews Immunology. 2020;20:217–228. [DOI] [PubMed] [Google Scholar]
  • 3.Chu C, Artis D and Chiu IM. Neuro-immune Interactions in the Tissues. Immunity. 2020;52:464–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Baral P, Udit S and Chiu IM. Pain and immunity: implications for host defence. Nature reviews Immunology. 2019;19:433–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zych AD and Gogolla N. Expressions of emotions across species. Curr Opin Neurobiol. 2021;68:57–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gogolla N The brain remembers where and how inflammation struck. Cell. 2021;184:5851–5853. [DOI] [PubMed] [Google Scholar]
  • 7.Mohanta SK, Weber C, Yin C and Habenicht AJR. The dawn has come for new therapeutics to treat atherosclerosis: Targeting neuroimmune cardiovascular interfaces in artery brain circuits. Clin Transl Med. 2022;12:e1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mohanta SK, Yin C, Weber C and Habenicht AJR. Neuroimmune cardiovascular interfaces in atherosclerosis. Front Cell Dev Biol. 2023;11:1117368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mohanta SK, Yin C, Peng L, Srikakulapu P, Bontha V, Hu D, Weih F, Weber C, Gerdes N and Habenicht AJ. Artery Tertiary Lymphoid Organs Contribute to Innate and Adaptive Immune Responses in Advanced Mouse Atherosclerosis. Circulation research. 2014;114:1772–1787. [DOI] [PubMed] [Google Scholar]
  • 10.Swirski FK and Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science. 2013;339:161–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dubin AE and Patapoutian A. Nociceptors: the sensors of the pain pathway. The Journal of clinical investigation. 2010;120:3760–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Patapoutian A, Tate S and Woolf CJ. Transient receptor potential channels: targeting pain at the source. Nature reviews Drug discovery. 2009;8:55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Tam SJ and Watts RJ. Connecting vascular and nervous system development: angiogenesis and the blood-brain barrier. Annual review of neuroscience. 2010;33:379–408. [DOI] [PubMed] [Google Scholar]
  • 14.Chiu IM, von Hehn CA and Woolf CJ. Neurogenic inflammation and the peripheral nervous system in host defense and immunopathology. Nature neuroscience. 2012;15:1063–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Talbot S, Foster SL and Woolf CJ. Neuroimmunity: Physiology and Pathology. Annual review of immunology. 2016;34:421–47. [DOI] [PubMed] [Google Scholar]
  • 16.Andersson U and Tracey KJ. Reflex principles of immunological homeostasis. Annual review of immunology. 2012;30:313–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Carmeliet P and Tessier-Lavigne M. Common mechanisms of nerve and blood vessel wiring. Nature. 2005;436:193–200. [DOI] [PubMed] [Google Scholar]
  • 18.McMahon SB, La Russa F and Bennett DL. Crosstalk between the nociceptive and immune systems in host defence and disease. Nature reviews Neuroscience. 2015;16:389–402. [DOI] [PubMed] [Google Scholar]
  • 19.Morrison SF and Nakamura K. Central Mechanisms for Thermoregulation. Annu Rev Physiol. 2019;81:285–308. [DOI] [PubMed] [Google Scholar]
  • 20.Pavlov VA, Chavan SS and Tracey KJ. Molecular and Functional Neuroscience in Immunity. Annual review of immunology. 2018;36:783–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pavlov VA and Tracey KJ. Neural regulation of immunity: molecular mechanisms and clinical translation. Nature neuroscience. 2017;20:156–166. [DOI] [PubMed] [Google Scholar]
  • 22.Chavan SS, Pavlov VA and Tracey KJ. Mechanisms and Therapeutic Relevance of Neuro-immune Communication. Immunity. 2017;46:927–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tracey KJ. Reflexes in Immunity. Cell. 2016;164:343–4. [DOI] [PubMed] [Google Scholar]
  • 24.O’Brien CJO, Haberman ER and Domingos AI. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu Rev Cell Dev Biol. 2021;37:549–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Schiller M, Ben-Shaanan TL and Rolls A. Neuronal regulation of immunity: why, how and where? Nature reviews Immunology. 2021;21:20–36. [DOI] [PubMed] [Google Scholar]
  • 26.Prescott SL and Liberles SD. Internal senses of the vagus nerve. Neuron. 2022;110:579–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Umans BD and Liberles SD. Neural Sensing of Organ Volume. Trends Neurosci. 2018;41:911–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Udit S, Blake K and Chiu IM. Somatosensory and autonomic neuronal regulation of the immune response. Nature reviews Neuroscience. 2022;23:157–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.You Z, Liu B and Qi H. Neuronal regulation of B-cell immunity: Anticipatory immune posturing? Neuron. 2022;110:3582–3596. [DOI] [PubMed] [Google Scholar]
  • 30.Wake E and Brack K. Characterization of the intrinsic cardiac nervous system. Autonomic neuroscience : basic & clinical. 2016;199:3–16. [DOI] [PubMed] [Google Scholar]
  • 31.Walchli T, Wacker A, Frei K, Regli L, Schwab ME, Hoerstrup SP, Gerhardt H and Engelhardt B. Wiring the Vascular Network with Neural Cues: A CNS Perspective. Neuron. 2015;87:271–96. [DOI] [PubMed] [Google Scholar]
  • 32.Haberman ER and Domingos AI. Illuminating Neuroimmunity: A Humoral Brain. Immunity. 2020;52:900–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Spector JM, Harrison RS and Fishman MC. Fundamental science behind today’s important medicines. Science translational medicine. 2018;10. [DOI] [PubMed] [Google Scholar]
  • 34.Fishman MC. Power of rare diseases: found in translation. Science translational medicine. 2013;5:201ps11. [DOI] [PubMed] [Google Scholar]
  • 35.Chen JN and Fishman MC. Genetics of heart development. Trends Genet. 2000;16:383–8. [DOI] [PubMed] [Google Scholar]
  • 36.Hsueh B, Chen R, Jo Y, Tang D, Raffiee M, Kim YS, Inoue M, Randles S, Ramakrishnan C, Patel S, Kim DK, Liu TX, Kim SH, Tan L, Mortazavi L, Cordero A, Shi J, Zhao M, Ho TT, Crow A, Yoo AW, Raja C, Evans K, Bernstein D, Zeineh M, Goubran M and Deisseroth K. Cardiogenic control of affective behavioural state. Nature. 2023;615:292–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Magnon C, Hall SJ, Lin J, Xue X, Gerber L, Freedland SJ and Frenette PS. Autonomic nerve development contributes to prostate cancer progression. Science. 2013;341:1236361. [DOI] [PubMed] [Google Scholar]
  • 38.Zeng W, Pirzgalska RM, Pereira MM, Kubasova N, Barateiro A, Seixas E, Lu YH, Kozlova A, Voss H, Martins GG, Friedman JM and Domingos AI. Sympathetic neuro-adipose connections mediate leptin-driven lipolysis. Cell. 2015;163:84–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Baral P, Umans BD, Li L, Wallrapp A, Bist M, Kirschbaum T, Wei Y, Zhou Y, Kuchroo VK, Burkett PR, Yipp BG, Liberles SD and Chiu IM. Nociceptor sensory neurons suppress neutrophil and gammadelta T cell responses in bacterial lung infections and lethal pneumonia. Nature medicine. 2018;24:417–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nonomura K, Woo SH, Chang RB, Gillich A, Qiu Z, Francisco AG, Ranade SS, Liberles SD and Patapoutian A. Piezo2 senses airway stretch and mediates lung inflation-induced apnoea. Nature. 2017;541:176–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tracey KJ. Reflex control of immunity. Nature reviews Immunology. 2009;9:418–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pongratz G and Straub RH. Role of peripheral nerve fibres in acute and chronic inflammation in arthritis. Nature reviews Rheumatology. 2013;9:117–26. [DOI] [PubMed] [Google Scholar]
  • 43.Klein AS, Dolensek N, Weiand C and Gogolla N. Fear balance is maintained by bodily feedback to the insular cortex in mice. Science. 2021;374:1010–1015. [DOI] [PubMed] [Google Scholar]
  • 44.Dolensek N, Gehrlach DA, Klein AS and Gogolla N. Facial expressions of emotion states and their neuronal correlates in mice. Science. 2020;368:89–94. [DOI] [PubMed] [Google Scholar]
  • 45.Moore KJ, Sheedy FJ and Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nature reviews Immunology. 2013;13:709–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kwon D Your brain could be controlling how sick you get - and how you recover. Nature. 2023;614:613–615. [DOI] [PubMed] [Google Scholar]
  • 47.Bin NR, Prescott SL, Horio N, Wang Y, Chiu IM and Liberles SD. An airway-to-brain sensory pathway mediates influenza-induced sickness. Nature. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bernhardt BC and Singer T. The neural basis of empathy. Annual review of neuroscience. 2012;35:1–23. [DOI] [PubMed] [Google Scholar]
  • 49.Gehrlach DA, Dolensek N, Klein AS, Roy Chowdhury R, Matthys A, Junghanel M, Gaitanos TN, Podgornik A, Black TD, Reddy Vaka N, Conzelmann KK and Gogolla N. Aversive state processing in the posterior insular cortex. Nature neuroscience. 2019;22:1424–1437. [DOI] [PubMed] [Google Scholar]
  • 50.Zhang X, Lei B, Yuan Y, Zhang L, Hu L, Jin S, Kang B, Liao X, Sun W, Xu F, Zhong Y, Hu J and Qi H. Brain control of humoral immune responses amenable to behavioural modulation. Nature. 2020;581:204–208. [DOI] [PubMed] [Google Scholar]
  • 51.Lai NY, Musser MA, Pinho-Ribeiro FA, Baral P, Jacobson A, Ma P, Potts DE, Chen Z, Paik D, Soualhi S, Yan Y, Misra A, Goldstein K, Lagomarsino VN, Nordstrom A, Sivanathan KN, Wallrapp A, Kuchroo VK, Nowarski R, Starnbach MN, Shi H, Surana NK, An D, Wu C, Huh JR, Rao M and Chiu IM. Gut-Innervating Nociceptor Neurons Regulate Peyer’s Patch Microfold Cells and SFB Levels to Mediate Salmonella Host Defense. Cell. 2020;180:33–49 e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Min S, Chang RB, Prescott SL, Beeler B, Joshi NR, Strochlic DE and Liberles SD. Arterial Baroreceptors Sense Blood Pressure through Decorated Aortic Claws. Cell Rep. 2019;29:2192–2201 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zeng WZ, Marshall KL, Min S, Daou I, Chapleau MW, Abboud FM, Liberles SD and Patapoutian A. PIEZOs mediate neuronal sensing of blood pressure and the baroreceptor reflex. Science. 2018;362:464–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mohanta SK, Peng L, Li Y, Lu S, Sun T, Carnevale L, Perrotta M, Ma Z, Forstera B, Stanic K, Zhang C, Zhang X, Szczepaniak P, Bianchini M, Saeed BR, Carnevale R, Hu D, Nosalski R, Pallante F, Beer M, Santovito D, Erturk A, Mettenleiter TC, Klupp BG, Megens RTA, Steffens S, Pelisek J, Eckstein HH, Kleemann R, Habenicht L, Mallat Z, Michel JB, Bernhagen J, Dichgans M, D’Agostino G, Guzik TJ, Olofsson PS, Yin C, Weber C, Lembo G, Carnevale D and Habenicht AJR. Neuroimmune cardiovascular interfaces control atherosclerosis. Nature. 2022;605:152–159. [DOI] [PubMed] [Google Scholar]
  • 55.Zhao Q, Yu CD, Wang R, Xu QJ, Dai Pra R, Zhang L and Chang RB. A multidimensional coding architecture of the vagal interoceptive system. Nature. 2022;603:878–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Libby P The changing landscape of atherosclerosis. Nature. 2021;592:524–533. [DOI] [PubMed] [Google Scholar]
  • 57.Libby P and Hansson GK. Inflammation and immunity in diseases of the arterial tree: players and layers. Circulation research. 2015;116:307–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Libby P, Ridker PM and Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011;473:317–25. [DOI] [PubMed] [Google Scholar]
  • 59.Weber C and Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nature medicine. 2011;17:1410–22. [DOI] [PubMed] [Google Scholar]
  • 60.Tabas I, Garcia-Cardena G and Owens GK. Recent insights into the cellular biology of atherosclerosis. The Journal of cell biology. 2015;209:13–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. The New England journal of medicine. 2005;352:1685–95. [DOI] [PubMed] [Google Scholar]
  • 62.Hansson GK. Inflammation, protection, and the problems of translation. Nat Rev Cardiol. 2018;15:729–730. [DOI] [PubMed] [Google Scholar]
  • 63.Zhao TX and Mallat Z. Targeting the Immune System in Atherosclerosis: JACC State-of-the-Art Review. Journal of the American College of Cardiology. 2019;73:1691–1706. [DOI] [PubMed] [Google Scholar]
  • 64.Mallat Z and Binder CJ. The why and how of adaptive immune responses in ischemic cardiovascular disease. Nat Cardiovasc Res. 2022;1:431–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Williams JW, Winkels H, Durant CP, Zaitsev K, Ghosheh Y and Ley K. Single Cell RNA Sequencing in Atherosclerosis Research. Circulation research. 2020;126:1112–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Winkels H, Gerdes N, Kahles F, Dutta P, Giannarelli C and Wolf D. Editorial: Immune and autoimmune mechanisms in cardiovascular disease. Front Cardiovasc Med. 2022;9:1125959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zhao L, Moos MP, Grabner R, Pedrono F, Fan J, Kaiser B, John N, Schmidt S, Spanbroek R, Lotzer K, Huang L, Cui J, Rader DJ, Evans JF, Habenicht AJ and Funk CD. The 5-lipoxygenase pathway promotes pathogenesis of hyperlipidemia-dependent aortic aneurysm. Nature medicine. 2004;10:966–73. [DOI] [PubMed] [Google Scholar]
  • 68.Moos MP, John N, Grabner R, Nossmann S, Gunther B, Vollandt R, Funk CD, Kaiser B and Habenicht AJ. The lamina adventitia is the major site of immune cell accumulation in standard chow-fed apolipoprotein E-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 2005;25:2386–91. [DOI] [PubMed] [Google Scholar]
  • 69.Grabner R, Lotzer K, Dopping S, Hildner M, Radke D, Beer M, Spanbroek R, Lippert B, Reardon CA, Getz GS, Fu YX, Hehlgans T, Mebius RE, van der Wall M, Kruspe D, Englert C, Lovas A, Hu D, Randolph GJ, Weih F and Habenicht AJ. Lymphotoxin beta receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE−/− mice. The Journal of experimental medicine. 2009;206:233–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Weih F, Grabner R, Hu D, Beer M and Habenicht AJ. Control of dichotomic innate and adaptive immune responses by artery tertiary lymphoid organs in atherosclerosis. Frontiers in physiology. 2012;3:226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hu D, Mohanta SK, Yin C, Peng L, Ma Z, Srikakulapu P, Grassia G, MacRitchie N, Dever G, Gordon P, Burton FL, Ialenti A, Sabir SR, McInnes IB, Brewer JM, Garside P, Weber C, Lehmann T, Teupser D, Habenicht L, Beer M, Grabner R, Maffia P, Weih F and Habenicht AJ. Artery Tertiary Lymphoid Organs Control Aorta Immunity and Protect against Atherosclerosis via Vascular Smooth Muscle Cell Lymphotoxin beta Receptors. Immunity. 2015;42:1100–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Srikakulapu P, Hu D, Yin C, Mohanta SK, Bontha SV, Peng L, Beer M, Weber C, McNamara CA, Grassia G, Maffia P, Manz RA and Habenicht AJ. Artery Tertiary Lymphoid Organs Control Multilayered Territorialized Atherosclerosis B-Cell Responses in Aged ApoE−/− Mice. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1174–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Yin C, Mohanta SK, Srikakulapu P, Weber C and Habenicht AJ. Artery Tertiary Lymphoid Organs: Powerhouses of Atherosclerosis Immunity. Frontiers in immunology. 2016;7:387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hu D, Yin C, Luo S, Habenicht AJR and Mohanta SK. Vascular Smooth Muscle Cells Contribute to Atherosclerosis Immunity. Frontiers in immunology. 2019;10:1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Akhavanpoor M, Gleissner CA, Akhavanpoor H, Lasitschka F, Doesch AO, Katus HA and Erbel C. Adventitial tertiary lymphoid organ classification in human atherosclerosis. Cardiovasc Pathol. 2018;32:8–14. [DOI] [PubMed] [Google Scholar]
  • 76.Yin C, Mohanta S, Maffia P and Habenicht AJ. Editorial: Tertiary Lymphoid Organs (TLOs): Powerhouses of Disease Immunity. Frontiers in immunology. 2017;8:228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Michel JB, Lagrange J, Regnault V and Lacolley P. Conductance Artery Wall Layers and Their Respective Roles in the Clearance Functions. Arteriosclerosis, thrombosis, and vascular biology. 2022;42:e253–e272. [DOI] [PubMed] [Google Scholar]
  • 78.Majesky MW. Adventitia and perivascular cells. Arteriosclerosis, thrombosis, and vascular biology. 2015;35:e31–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Majesky MW and Weiser-Evans MCM. The adventitia in arterial development, remodeling, and hypertension. Biochemical pharmacology. 2022;205:115259. [DOI] [PubMed] [Google Scholar]
  • 80.Andersson U and Tracey KJ. Neural reflexes in inflammation and immunity. The Journal of experimental medicine. 2012;209:1057–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Huang S, Ziegler CGK, Austin J, Mannoun N, Vukovic M, Ordovas-Montanes J, Shalek AK and von Andrian UH. Lymph nodes are innervated by a unique population of sensory neurons with immunomodulatory potential. Cell. 2021;184:441–459 e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Julius D and Basbaum AI. Molecular mechanisms of nociception. Nature. 2001;413:203–10. [DOI] [PubMed] [Google Scholar]
  • 83.Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD and Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–24. [DOI] [PubMed] [Google Scholar]
  • 84.Patapoutian A and Macpherson L. Channeling pain. Nature medicine. 2006;12:506–7. [DOI] [PubMed] [Google Scholar]
  • 85.Hinterdobler J, Schott S, Jin H, Meesmann A, Steinsiek AL, Zimmermann AS, Wobst J, Muller P, Mauersberger C, Vilne B, Baecklund A, Chen CS, Moggio A, Braster Q, Molitor M, Krane M, Kempf WE, Ladwig KH, Hristov M, Hulsmans M, Hilgendorf I, Weber C, Wenzel P, Scheiermann C, Maegdefessel L, Soehnlein O, Libby P, Nahrendorf M, Schunkert H, Kessler T and Sager HB. Acute mental stress drives vascular inflammation and promotes plaque destabilization in mouse atherosclerosis. European heart journal. 2021;42:4077–4088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Armour JA. Potential clinical relevance of the ‘little brain’ on the mammalian heart. Experimental physiology. 2008;93:165–76. [DOI] [PubMed] [Google Scholar]
  • 87.Berthoud HR and Neuhuber WL. Functional and chemical anatomy of the afferent vagal system. Autonomic neuroscience : basic & clinical. 2000;85:1–17. [DOI] [PubMed] [Google Scholar]
  • 88.Meltzer S, Santiago C, Sharma N and Ginty DD. The cellular and molecular basis of somatosensory neuron development. Neuron. 2021;109:3736–3757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Paintal AS. Vagal sensory receptors and their reflex effects. Physiol Rev. 1973;53:159–227. [DOI] [PubMed] [Google Scholar]
  • 90.Hainsworth R Reflexes from the heart. Physiological reviews. 1991;71:617–58. [DOI] [PubMed] [Google Scholar]
  • 91.Kaufman MP, Baker DG, Coleridge HM and Coleridge JC. Stimulation by bradykinin of afferent vagal C-fibers with chemosensitive endings in the heart and aorta of the dog. Circ Res. 1980;46:476–84. [DOI] [PubMed] [Google Scholar]
  • 92.Cheng Z, Powley TL, Schwaber JS and Doyle FJ 3rd. A laser confocal microscopic study of vagal afferent innervation of rat aortic arch: chemoreceptors as well as baroreceptors. Journal of the autonomic nervous system. 1997;67:1–14. [DOI] [PubMed] [Google Scholar]
  • 93.Cheng Z, Powley TL, Schwaber JS and Doyle FJ 3rd. Vagal afferent innervation of the atria of the rat heart reconstructed with confocal microscopy. J Comp Neurol. 1997;381:1–17. [DOI] [PubMed] [Google Scholar]
  • 94.Crystal GJ and Salem MR. The Bainbridge and the “reverse” Bainbridge reflexes: history, physiology, and clinical relevance. Anesth Analg. 2012;114:520–32. [DOI] [PubMed] [Google Scholar]
  • 95.Aviado DM and Guevara Aviado D. The Bezold-Jarisch reflex. A historical perspective of cardiopulmonary reflexes. Ann N Y Acad Sci. 2001;940:48–58. [PubMed] [Google Scholar]
  • 96.Campagna JA and Carter C. Clinical relevance of the Bezold-Jarisch reflex. Anesthesiology. 2003;98:1250–60. [DOI] [PubMed] [Google Scholar]
  • 97.Lu Y, Ma X, Sabharwal R, Snitsarev V, Morgan D, Rahmouni K, Drummond HA, Whiteis CA, Costa V, Price M, Benson C, Welsh MJ, Chapleau MW and Abboud FM. The ion channel ASIC2 is required for baroreceptor and autonomic control of the circulation. Neuron. 2009;64:885–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lau OC, Shen B, Wong CO, Tjong YW, Lo CY, Wang HC, Huang Y, Yung WH, Chen YC, Fung ML, Rudd JA and Yao X. TRPC5 channels participate in pressure-sensing in aortic baroreceptors. Nat Commun. 2016;7:11947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lu HJ, Nguyen TL, Hong GS, Pak S, Kim H, Kim H, Kim DY, Kim SY, Shen Y, Ryu PD, Lee MO and Oh U. Tentonin 3/TMEM150C senses blood pressure changes in the aortic arch. The Journal of clinical investigation. 2020;130:3671–3683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chen WW, Xiong XQ, Chen Q, Li YH, Kang YM and Zhu GQ. Cardiac sympathetic afferent reflex and its implications for sympathetic activation in chronic heart failure and hypertension. Acta Physiol (Oxf). 2015;213:778–94. [DOI] [PubMed] [Google Scholar]
  • 101.Gao L, Zhu Z, Zucker IH and Wang W. Cardiac sympathetic afferent stimulation impairs baroreflex control of renal sympathetic nerve activity in rats. Am J Physiol Heart Circ Physiol. 2004;286:H1706–11. [DOI] [PubMed] [Google Scholar]
  • 102.Wink J, van Delft R, Notenboom RGE, Wouters PF, DeRuiter MC, Plevier JWM and Jongbloed MRM. Human adult cardiac autonomic innervation: Controversies in anatomical knowledge and relevance for cardiac neuromodulation. Auton Neurosci. 2020;227:102674. [DOI] [PubMed] [Google Scholar]
  • 103.Tao J, Campbell JN, Tsai LT, Wu C, Liberles SD and Lowell BB. Highly selective brain-to-gut communication via genetically defined vagus neurons. Neuron. 2021;109:2106–2115 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fedele L and Brand T. The Intrinsic Cardiac Nervous System and Its Role in Cardiac Pacemaking and Conduction. J Cardiovasc Dev Dis. 2020;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.MacDonald EA, Rose RA and Quinn TA. Neurohumoral Control of Sinoatrial Node Activity and Heart Rate: Insight From Experimental Models and Findings From Humans. Front Physiol. 2020;11:170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gardner RT, Ripplinger CM, Myles RC and Habecker BA. Molecular Mechanisms of Sympathetic Remodeling and Arrhythmias. Circ Arrhythm Electrophysiol. 2016;9:e001359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Duraes Campos I, Pinto V, Sousa N and Pereira VH. A brain within the heart: A review on the intracardiac nervous system. J Mol Cell Cardiol. 2018;119:1–9. [DOI] [PubMed] [Google Scholar]
  • 108.Pauza DH, Skripka V, Pauziene N and Stropus R. Morphology, distribution, and variability of the epicardiac neural ganglionated subplexuses in the human heart. Anat Rec. 2000;259:353–82. [DOI] [PubMed] [Google Scholar]
  • 109.Gagliardi M, Randall WC, Bieger D, Wurster RD, Hopkins DA and Armour JA. Activity of in vivo canine cardiac plexus neurons. Am J Physiol. 1988;255:H789–800. [DOI] [PubMed] [Google Scholar]
  • 110.Chan KY, Jang MJ, Yoo BB, Greenbaum A, Ravi N, Wu WL, Sanchez-Guardado L, Lois C, Mazmanian SK, Deverman BE and Gradinaru V. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat Neurosci. 2017;20:1172–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Rajendran PS, Challis RC, Fowlkes CC, Hanna P, Tompkins JD, Jordan MC, Hiyari S, Gabris-Weber BA, Greenbaum A, Chan KY, Deverman BE, Munzberg H, Ardell JL, Salama G, Gradinaru V and Shivkumar K. Identification of peripheral neural circuits that regulate heart rate using optogenetic and viral vector strategies. Nature communications. 2019;10:1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Saburkina I, Gukauskiene L, Rysevaite K, Brack KE, Pauza AG, Pauziene N and Pauza DH. Morphological pattern of intrinsic nerve plexus distributed on the rabbit heart and interatrial septum. J Anat. 2014;224:583–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lizot G, Pasqualin C, Tissot A, Pages S, Faivre JF and Chatelier A. Molecular and functional characterization of the mouse intrinsic cardiac nervous system. Heart Rhythm. 2022;19:1352–1362. [DOI] [PubMed] [Google Scholar]
  • 114.Veerakumar A, Yung AR, Liu Y and Krasnow MA. Molecularly defined circuits for cardiovascular and cardiopulmonary control. Nature. 2022;606:739–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hanna P, Dacey MJ, Brennan J, Moss A, Robbins S, Achanta S, Biscola NP, Swid MA, Rajendran PS, Mori S, Hadaya JE, Smith EH, Peirce SG, Chen J, Havton LA, Cheng ZJ, Vadigepalli R, Schwaber J, Lux RL, Efimov I, Tompkins JD, Hoover DB, Ardell JL and Shivkumar K. Innervation and Neuronal Control of the Mammalian Sinoatrial Node a Comprehensive Atlas. Circ Res. 2021;128:1279–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Herring N, Lokale MN, Danson EJ, Heaton DA and Paterson DJ. Neuropeptide Y reduces acetylcholine release and vagal bradycardia via a Y2 receptor-mediated, protein kinase C-dependent pathway. J Mol Cell Cardiol. 2008;44:477–85. [DOI] [PubMed] [Google Scholar]
  • 117.Achanta S, Gorky J, Leung C, Moss A, Robbins S, Eisenman L, Chen J, Tappan S, Heal M, Farahani N, Huffman T, England S, Cheng ZJ, Vadigepalli R and Schwaber JS. A Comprehensive Integrated Anatomical and Molecular Atlas of Rat Intrinsic Cardiac Nervous System. iScience. 2020;23:101140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Rimmer K and Harper AA. Developmental changes in electrophysiological properties and synaptic transmission in rat intracardiac ganglion neurons. J Neurophysiol. 2006;95:3543–52. [DOI] [PubMed] [Google Scholar]
  • 119.Edwards FR, Hirst GD, Klemm MF and Steele PA. Different types of ganglion cell in the cardiac plexus of guinea-pigs. J Physiol. 1995;486 (Pt 2):453–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Klemm MF, Wallace DJ and Hirst GD. Distribution of synaptic boutons around identified neurones lying in the cardiac plexus of the guinea-pig. J Auton Nerv Syst. 1997;66:201–7. [DOI] [PubMed] [Google Scholar]
  • 121.Allen E, Coote JH, Grubb BD, Batten TFC, Pauza DH, Ng GA and Brack KE. Electrophysiological effects of nicotinic and electrical stimulation of intrinsic cardiac ganglia in the absence of extrinsic autonomic nerves in the rabbit heart. Heart Rhythm. 2018;15:1698–1707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.McAllen RM, Salo LM, Paton JF and Pickering AE. Processing of central and reflex vagal drives by rat cardiac ganglion neurones: an intracellular analysis. J Physiol. 2011;589:5801–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Beaumont E, Salavatian S, Southerland EM, Vinet A, Jacquemet V, Armour JA and Ardell JL. Network interactions within the canine intrinsic cardiac nervous system: implications for reflex control of regional cardiac function. J Physiol. 2013;591:4515–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Shen MJ and Zipes DP. Role of the autonomic nervous system in modulating cardiac arrhythmias. Circ Res. 2014;114:1004–21. [DOI] [PubMed] [Google Scholar]
  • 125.Choi EK, Shen MJ, Han S, Kim D, Hwang S, Sayfo S, Piccirillo G, Frick K, Fishbein MC, Hwang C, Lin SF and Chen PS. Intrinsic cardiac nerve activity and paroxysmal atrial tachyarrhythmia in ambulatory dogs. Circulation. 2010;121:2615–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Pappone C, Santinelli V, Manguso F, Vicedomini G, Gugliotta F, Augello G, Mazzone P, Tortoriello V, Landoni G, Zangrillo A, Lang C, Tomita T, Mesas C, Mastella E and Alfieri O. Pulmonary vein denervation enhances long-term benefit after circumferential ablation for paroxysmal atrial fibrillation. Circulation. 2004;109:327–34. [DOI] [PubMed] [Google Scholar]
  • 127.Jungen C, Scherschel K, Eickholt C, Kuklik P, Klatt N, Bork N, Salzbrunn T, Alken F, Angendohr S, Klene C, Mester J, Klocker N, Veldkamp MW, Schumacher U, Willems S, Nikolaev VO and Meyer C. Disruption of cardiac cholinergic neurons enhances susceptibility to ventricular arrhythmias. Nat Commun. 2017;8:14155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Huang MH, Wolf SG and Armour JA. Ventricular arrhythmias induced by chemically modified intrinsic cardiac neurones. Cardiovasc Res. 1994;28:636–42. [DOI] [PubMed] [Google Scholar]
  • 129.Swirski FK and Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nature reviews Immunology. 2018;18:733–744. [DOI] [PubMed] [Google Scholar]
  • 130.Koc A and Cagavi E. Cardiac Immunology: A New Era for Immune Cells in the Heart. Advances in experimental medicine and biology. 2021;1312:75–95. [DOI] [PubMed] [Google Scholar]
  • 131.Poller WC, Nahrendorf M and Swirski FK. Hematopoiesis and Cardiovascular Disease. Circulation research. 2020;126:1061–1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Sauter M and Langer HF. Targeting Cell-Specific Molecular Mechanisms of Innate Immunity in Atherosclerosis. Frontiers in physiology. 2022;13:802990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wang Z, Zhang X, Lu S, Zhang Chuankai, Ma Zhe, Su Rui, Li Yuanfang, Sun Ting, Li Yutao, Hong Mingyang, Deng Xinyi, Monjezi Mohammad Rafiee, Hristov Michael, Steffens Sabine, Santovito Donato, Dornmair Klaus, Ley Klaus, Weber Christian, Mohanta Sarajo K., Habenicht Andreas, Yin Changjun. Pairing of single-cell RNA analysis and T-cell receptor profiling indicates breakdown of T-cell tolerance checkpoints in atherosclerosis. Nature Cardiovascular Research. 2023; 2: 290–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Medzhitov R The spectrum of inflammatory responses. Science. 2021;374:1070–1075. [DOI] [PubMed] [Google Scholar]
  • 135.Wang A, Luan HH and Medzhitov R. An evolutionary perspective on immunometabolism. Science. 2019;363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lorenzo C, Delgado P, Busse CE, Sanz-Bravo A, Martos-Folgado I, Bonzon-Kulichenko E, Ferrarini A, Gonzalez-Valdes IB, Mur SM, Roldan-Montero R, Martinez-Lopez D, Martin-Ventura JL, Vazquez J, Wardemann H and Ramiro AR. ALDH4A1 is an atherosclerosis auto-antigen targeted by protective antibodies. Nature. 2021;589:287–292. [DOI] [PubMed] [Google Scholar]
  • 137.Edsfeldt A, Nilsson J Understanding autoimmunity in atherosclerosis paves the way for novel therapies. Nature Cardiovascular Research. 2023;2:227–229. [DOI] [PubMed] [Google Scholar]
  • 138.Depuydt MAC, Schaftenaar FH, Prange KHM, Boltjes A, Hemme E, Delfos L, de Mol J, de Jong MJM, Kleijn MNAB, Peeters JAHM, Goncalves L, Wezel A, Smeets JH, de Borst GJ, Foks AC, Pasterkamp G, de Winther MPJ, Kuiper J, Bot I, Slütter B Single-cell T cell receptor sequencing of paired human atherosclerotic plaques and blood reveals autoimmune-like features of expanded effector T cells. Nature Cardiovascular Research. 2023;2:112–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Parra Sanchez AR, Voskuyl AE and van Vollenhoven RF. Treat-to-target in systemic lupus erythematosus: advancing towards its implementation. Nature reviews Rheumatology. 2022;18:146–157. [DOI] [PubMed] [Google Scholar]
  • 140.Ding Q, Hu W, Wang R, Yang Q, Zhu M, Li M, Cai J, Rose P, Mao J and Zhu YZ. Signaling pathways in rheumatoid arthritis: implications for targeted therapy. Signal Transduct Target Ther. 2023;8:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Caielli S, Wan Z and Pascual V. Systemic Lupus Erythematosus Pathogenesis: Interferon and Beyond. Annual review of immunology. 2023;41:533–560. [DOI] [PubMed] [Google Scholar]
  • 142.Li ZY, Cai ML, Qin Y and Chen Z. Age/autoimmunity-associated B cells in inflammatory arthritis: An emerging therapeutic target. Frontiers in immunology. 2023;14:1103307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Godinho-Silva C, Cardoso F and Veiga-Fernandes H. Neuro-Immune Cell Units: A New Paradigm in Physiology. Annual review of immunology. 2019;37:19–46. [DOI] [PubMed] [Google Scholar]
  • 144.Chesne J, Cardoso V and Veiga-Fernandes H. Neuro-immune regulation of mucosal physiology. Mucosal immunology. 2019;12:10–20. [DOI] [PubMed] [Google Scholar]
  • 145.Muller PA, Koscso B, Rajani GM, Stevanovic K, Berres ML, Hashimoto D, Mortha A, Leboeuf M, Li XM, Mucida D, Stanley ER, Dahan S, Margolis KG, Gershon MD, Merad M and Bogunovic M. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell. 2014;158:300–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gabanyi I, Muller PA, Feighery L, Oliveira TY, Costa-Pinto FA and Mucida D. Neuro-immune Interactions Drive Tissue Programming in Intestinal Macrophages. Cell. 2016;164:378–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Matheis F, Muller PA, Graves CL, Gabanyi I, Kerner ZJ, Costa-Borges D, Ahrends T, Rosenstiel P and Mucida D. Adrenergic Signaling in Muscularis Macrophages Limits Infection-Induced Neuronal Loss. Cell. 2020;180:64–78 e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Pirzgalska RM, Seixas E, Seidman JS, Link VM, Sanchez NM, Mahu I, Mendes R, Gres V, Kubasova N, Morris I, Arus BA, Larabee CM, Vasques M, Tortosa F, Sousa AL, Anandan S, Tranfield E, Hahn MK, Iannacone M, Spann NJ, Glass CK and Domingos AI. Sympathetic neuron-associated macrophages contribute to obesity by importing and metabolizing norepinephrine. Nature medicine. 2017;23:1309–1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Camell CD, Sander J, Spadaro O, Lee A, Nguyen KY, Wing A, Goldberg EL, Youm YH, Brown CW, Elsworth J, Rodeheffer MS, Schultze JL and Dixit VD. Inflammasome-driven catecholamine catabolism in macrophages blunts lipolysis during ageing. Nature. 2017;550:119–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Wolf D, Gerhardt T, Winkels H, Michel NA, Pramod AB, Ghosheh Y, Brunel S, Buscher K, Miller J, McArdle S, Baas L, Kobiyama K, Vassallo M, Ehinger E, Dileepan T, Ali A, Schell M, Mikulski Z, Sidler D, Kimura T, Sheng X, Horstmann H, Hansen S, Mitre LS, Stachon P, Hilgendorf I, Gaddis DE, Hedrick C, Benedict CA, Peters B, Zirlik A, Sette A and Ley K. Pathogenic Autoimmunity in Atherosclerosis Evolves From Initially Protective Apolipoprotein B100-Reactive CD4(+) T-Regulatory Cells. Circulation. 2020;142:1279–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.van Gils JM, Derby MC, Fernandes LR, Ramkhelawon B, Ray TD, Rayner KJ, Parathath S, Distel E, Feig JL, Alvarez-Leite JI, Rayner AJ, McDonald TO, O’Brien KD, Stuart LM, Fisher EA, Lacy-Hulbert A and Moore KJ. The neuroimmune guidance cue netrin-1 promotes atherosclerosis by inhibiting the emigration of macrophages from plaques. Nature immunology. 2012;13:136–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Ibiza S, Garcia-Cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, Misic AM, Bartow-McKenney C, Larson DM, Pavan WJ, Eberl G, Grice EA and Veiga-Fernandes H. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature. 2016;535:440–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Cardoso V, Chesne J, Ribeiro H, Garcia-Cassani B, Carvalho T, Bouchery T, Shah K, Barbosa-Morais NL, Harris N and Veiga-Fernandes H. Neuronal regulation of type 2 innate lymphoid cells via neuromedin U. Nature. 2017;549:277–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour RE, Nyman J, Dionne D, Hofree M, Cuoco MS, Rodman C, Farouq D, Haas BJ, Tickle TL, Trombetta JJ, Baral P, Klose CSN, Mahlakoiv T, Artis D, Rozenblatt-Rosen O, Chiu IM, Levy BD, Kowalczyk MS, Regev A and Kuchroo VK. The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature. 2017;549:351–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Klose CSN, Mahlakoiv T, Moeller JB, Rankin LC, Flamar AL, Kabata H, Monticelli LA, Moriyama S, Putzel GG, Rakhilin N, Shen X, Kostenis E, Konig GM, Senda T, Carpenter D, Farber DL and Artis D. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature. 2017;549:282–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Moriyama S, Brestoff JR, Flamar AL, Moeller JB, Klose CSN, Rankin LC, Yudanin NA, Monticelli LA, Putzel GG, Rodewald HR and Artis D. beta(2)-adrenergic receptor-mediated negative regulation of group 2 innate lymphoid cell responses. Science. 2018;359:1056–1061. [DOI] [PubMed] [Google Scholar]
  • 157.Talbot S, Abdulnour RE, Burkett PR, Lee S, Cronin SJ, Pascal MA, Laedermann C, Foster SL, Tran JV, Lai N, Chiu IM, Ghasemlou N, DiBiase M, Roberson D, Von Hehn C, Agac B, Haworth O, Seki H, Penninger JM, Kuchroo VK, Bean BP, Levy BD and Woolf CJ. Silencing Nociceptor Neurons Reduces Allergic Airway Inflammation. Neuron. 2015;87:341–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Chen WY, Wu YH, Tsai TH, Li RF, Lai AC, Li LC, Yang JL and Chang YJ. Group 2 innate lymphoid cells contribute to IL-33-mediated alleviation of cardiac fibrosis. Theranostics. 2021;11:2594–2611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Yu X, Newland SA, Zhao TX, Lu Y, Sage AS, Sun Y, Sriranjan RS, Ma MKL, Lam BYH, Nus M, Harrison JE, Bond SJ, Cheng X, Silvestre JS, Rudd JHF, Cheriyan J and Mallat Z. Innate Lymphoid Cells Promote Recovery of Ventricular Function After Myocardial Infarction. Journal of the American College of Cardiology. 2021;78:1127–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Godinho-Silva C, Domingues RG, Rendas M, Raposo B, Ribeiro H, da Silva JA, Vieira A, Costa RM, Barbosa-Morais NL, Carvalho T and Veiga-Fernandes H. Light-entrained and brain-tuned circadian circuits regulate ILC3s and gut homeostasis. Nature. 2019;574:254–+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Cardoso F, Klein Wolterink RGJ, Godinho-Silva C, Domingues RG, Ribeiro H, da Silva JA, Mahu I, Domingos AI and Veiga-Fernandes H. Neuro-mesenchymal units control ILC2 and obesity via a brain-adipose circuit. Nature. 2021;597:410–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Poller WC, Downey J, Mooslechner AA, Khan N, Li L, Chan CT, McAlpine CS, Xu C, Kahles F, He S, Janssen H, Mindur JE, Singh S, Kiss MG, Alonso-Herranz L, Iwamoto Y, Kohler RH, Wong LP, Chetal K, Russo SJ, Sadreyev RI, Weissleder R, Nahrendorf M, Frenette PS, Divangahi M and Swirski FK. Brain motor and fear circuits regulate leukocytes during acute stress. Nature. 2022;607:578–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Koren T, Yifa R, Amer M, Krot M, Boshnak N, Ben-Shaanan TL, Azulay-Debby H, Zalayat I, Avishai E, Hajjo H, Schiller M, Haykin H, Korin B, Farfara D, Hakim F, Kobiler O, Rosenblum K and Rolls A. Insular cortex neurons encode and retrieve specific immune responses. Cell. 2021;184:5902–5915 e17. [DOI] [PubMed] [Google Scholar]
  • 164.Nahrendorf M and Swirski FK. Lifestyle effects on hematopoiesis and atherosclerosis. Circulation research. 2015;116:884–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Schloss MJ, Hulsmans M, Rohde D, Lee IH, Severe N, Foy BH, Pulous FE, Zhang S, Kokkaliaris KD, Frodermann V, Courties G, Yang C, Iwamoto Y, Knudsen AS, McAlpine CS, Yamazoe M, Schmidt SP, Wojtkiewicz GR, Masson GS, Gustafsson K, Capen D, Brown D, Higgins JM, Scadden DT, Libby P, Swirski FK, Naxerova K and Nahrendorf M. B lymphocyte-derived acetylcholine limits steady-state and emergency hematopoiesis. Nature immunology. 2022;23:605–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.McAlpine CS, Kiss MG, Zuraikat FM, Cheek D, Schiroli G, Amatullah H, Huynh P, Bhatti MZ, Wong LP, Yates AG, Poller WC, Mindur JE, Chan CT, Janssen H, Downey J, Singh S, Sadreyev RI, Nahrendorf M, Jeffrey KL, Scadden DT, Naxerova K, St-Onge MP and Swirski FK. Sleep exerts lasting effects on hematopoietic stem cell function and diversity. The Journal of experimental medicine. 2022;219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Barrett TJ, Corr EM, van Solingen C, Schlamp F, Brown EJ, Koelwyn GJ, Lee AH, Shanley LC, Spruill TM, Bozal F, de Jong A, Newman AAC, Drenkova K, Silvestro M, Ramkhelawon B, Reynolds HR, Hochman JS, Nahrendorf M, Swirski FK, Fisher EA, Berger JS and Moore KJ. Chronic stress primes innate immune responses in mice and humans. Cell Rep. 2021;36:109595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Mendez-Ferrer S, Lucas D, Battista M and Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–7. [DOI] [PubMed] [Google Scholar]
  • 169.Yvan-Charvet L, Pagler T, Gautier EL, Avagyan S, Siry RL, Han S, Welch CL, Wang N, Randolph GJ, Snoeck HW and Tall AR. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science. 2010;328:1689–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Gu Q, Yang X, Lv J, Zhang J, Xia B, Kim JD, Wang R, Xiong F, Meng S, Clements TP, Tandon B, Wagner DS, Diaz MF, Wenzel PL, Miller YI, Traver D, Cooke JP, Li W, Zon LI, Chen K, Bai Y and Fang L. AIBP-mediated cholesterol efflux instructs hematopoietic stem and progenitor cell fate. Science. 2019;363:1085–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Heidt T, Sager HB, Courties G, Dutta P, Iwamoto Y, Zaltsman A, von Zur Muhlen C, Bode C, Fricchione GL, Denninger J, Lin CP, Vinegoni C, Libby P, Swirski FK, Weissleder R and Nahrendorf M. Chronic variable stress activates hematopoietic stem cells. Nature medicine. 2014;20:754–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Frodermann V, Rohde D, Courties G, Severe N, Schloss MJ, Amatullah H, McAlpine CS, Cremer S, Hoyer FF, Ji F, van Koeverden ID, Herisson F, Honold L, Masson GS, Zhang S, Grune J, Iwamoto Y, Schmidt SP, Wojtkiewicz GR, Lee IH, Gustafsson K, Pasterkamp G, de Jager SCA, Sadreyev RI, MacFadyen J, Libby P, Ridker P, Scadden DT, Naxerova K, Jeffrey KL, Swirski FK and Nahrendorf M. Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells. Nature medicine. 2019;25:1761–1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.McAlpine CS, Kiss MG, Rattik S, He S, Vassalli A, Valet C, Anzai A, Chan CT, Mindur JE, Kahles F, Poller WC, Frodermann V, Fenn AM, Gregory AF, Halle L, Iwamoto Y, Hoyer FF, Binder CJ, Libby P, Tafti M, Scammell TE, Nahrendorf M and Swirski FK. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature. 2019;566:383–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Nagai M, Hoshide S and Kario K. The insular cortex and cardiovascular system: a new insight into the brain-heart axis. J Am Soc Hypertens. 2010;4:174–82. [DOI] [PubMed] [Google Scholar]
  • 175.Tawakol A, Ishai A, Takx RA, Figueroa AL, Ali A, Kaiser Y, Truong QA, Solomon CJ, Calcagno C, Mani V, Tang CY, Mulder WJ, Murrough JW, Hoffmann U, Nahrendorf M, Shin LM, Fayad ZA and Pitman RK. Relation between resting amygdalar activity and cardiovascular events: a longitudinal and cohort study. Lancet. 2017;389:834–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Schaeuble D and Myers B. Cortical-Hypothalamic Integration of Autonomic and Endocrine Stress Responses. Frontiers in physiology. 2022;13:820398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Manuel J, Farber N, Gerlach DA, Heusser K, Jordan J, Tank J and Beissner F. Deciphering the neural signature of human cardiovascular regulation. Elife. 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Davern PJ. A role for the lateral parabrachial nucleus in cardiovascular function and fluid homeostasis. Frontiers in physiology. 2014;5:436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kumagai H, Oshima N, Matsuura T, Iigaya K, Imai M, Onimaru H, Sakata K, Osaka M, Onami T, Takimoto C, Kamayachi T, Itoh H and Saruta T. Importance of rostral ventrolateral medulla neurons in determining efferent sympathetic nerve activity and blood pressure. Hypertens Res. 2012;35:132–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Karemaker JM. The multibranched nerve: vagal function beyond heart rate variability. Biol Psychol. 2022;172:108378. [DOI] [PubMed] [Google Scholar]
  • 181.Zanutto BS, Valentinuzzi ME and Segura ET. Neural set point for the control of arterial pressure: role of the nucleus tractus solitarius. Biomed Eng Online. 2010;9:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Carnevale D Neuroimmune axis of cardiovascular control: mechanisms and therapeutic implications. Nat Rev Cardiol. 2022;19:379–394. [DOI] [PubMed] [Google Scholar]
  • 183.Hu H Reward and Aversion. Annual review of neuroscience. 2016;39:297–324. [DOI] [PubMed] [Google Scholar]
  • 184.Hsueh B, Chen R, Jo Y, Tang D, Raffiee M, Kim YS, Inoue M, Randles S, Ramakrishnan C, Patel S, Kim DK, Liu TX, Kim SH, Tan L, Mortazavi L, Cordero A, Shi J, Zhao M, Ho TT, Crow A, Yoo AW, Raja C, Evans K, Bernstein D, Zeineh M, Goubran M and Deisseroth K. Cardiogenic control of affective behavioural state. Nature. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Josselyn SA and Frankland PW. Memory Allocation: Mechanisms and Function. Annual review of neuroscience. 2018;41:389–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Romanov RA, Alpar A, Hokfelt T and Harkany T. Unified Classification of Molecular, Network, and Endocrine Features of Hypothalamic Neurons. Annual review of neuroscience. 2019;42:1–26. [DOI] [PubMed] [Google Scholar]
  • 187.Ruder L and Arber S. Brainstem Circuits Controlling Action Diversification. Annual review of neuroscience. 2019;42:485–504. [DOI] [PubMed] [Google Scholar]
  • 188.Pavlov VA, Wang H, Czura CJ, Friedman SG and Tracey KJ. The cholinergic anti-inflammatory pathway: a missing link in neuroimmunomodulation. Mol Med. 2003;9:125–34. [PMC free article] [PubMed] [Google Scholar]
  • 189.Weinstein BM, Stemple DL, Driever W and Fishman MC. Gridlock, a localized heritable vascular patterning defect in the zebrafish. Nature medicine. 1995;1:1143–7. [DOI] [PubMed] [Google Scholar]
  • 190.Singh AP, Sosa MX, Fang J, Shanmukhappa SK, Hubaud A, Fawcett CH, Molind GJ, Tsai T, Capodieci P, Wetzel K, Sanchez E, Wang G, Coble M, Tang W, Cadena SM, Fishman MC and Glass DJ. alphaKlotho Regulates Age-Associated Vascular Calcification and Lifespan in Zebrafish. Cell Rep. 2019;28:2767–2776 e5. [DOI] [PubMed] [Google Scholar]
  • 191.Krause T, Werner K, Fiebach JB, Villringer K, Piper SK, Haeusler KG, Endres M, Scheitz JF and Nolte CH. Stroke in right dorsal anterior insular cortex Is related to myocardial injury. Annals of neurology. 2017;81:502–511. [DOI] [PubMed] [Google Scholar]
  • 192.Hansson GK and Libby P. The immune response in atherosclerosis: a double-edged sword. Nature reviews Immunology. 2006;6:508–19. [DOI] [PubMed] [Google Scholar]
  • 193.Gogolla N The insular cortex. Current biology : CB. 2017;27:R580–R586. [DOI] [PubMed] [Google Scholar]
  • 194.Schacter DL. Forgotten Ideas, Neglected Pioneers: Richard Semon and the Story of Memory: Psychology Press; 2001. [Google Scholar]
  • 195.Josselyn SA and Tonegawa S. Memory engrams: Recalling the past and imagining the future. Science. 2020;367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Brea D and Veiga-Fernandes H. Inflammation in the gut is encoded by neurons in the brain. Nature. 2022;602:217–218. [DOI] [PubMed] [Google Scholar]
  • 197.Willems T and Henke K. Imaging human engrams using 7 Tesla magnetic resonance imaging. Hippocampus. 2021;31:1257–1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Yin C, Ackermann S, Ma Z, Mohanta SK, Zhang C, Li Y, Nietzsche S, Westermann M, Peng L, Hu D, Bontha SV, Srikakulapu P, Beer M, Megens RTA, Steffens S, Hildner M, Halder LD, Eckstein HH, Pelisek J, Herms J, Roeber S, Arzberger T, Borodovsky A, Habenicht L, Binder CJ, Weber C, Zipfel PF, Skerka C and Habenicht AJR. ApoE attenuates unresolvable inflammation by complex formation with activated C1q. Nature medicine. 2019;25:496–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Bakken TE, Jorstad NL, Hu Q, Lake BB, Tian W, Kalmbach BE, Crow M, Hodge RD, Krienen FM, Sorensen SA, Eggermont J, Yao Z, Aevermann BD, Aldridge AI, Bartlett A, Bertagnolli D, Casper T, Castanon RG, Crichton K, Daigle TL, Dalley R, Dee N, Dembrow N, Diep D, Ding SL, Dong W, Fang R, Fischer S, Goldman M, Goldy J, Graybuck LT, Herb BR, Hou X, Kancherla J, Kroll M, Lathia K, van Lew B, Li YE, Liu CS, Liu H, Lucero JD, Mahurkar A, McMillen D, Miller JA, Moussa M, Nery JR, Nicovich PR, Niu SY, Orvis J, Osteen JK, Owen S, Palmer CR, Pham T, Plongthongkum N, Poirion O, Reed NM, Rimorin C, Rivkin A, Romanow WJ, Sedeno-Cortes AE, Siletti K, Somasundaram S, Sulc J, Tieu M, Torkelson A, Tung H, Wang X, Xie F, Yanny AM, Zhang R, Ament SA, Behrens MM, Bravo HC, Chun J, Dobin A, Gillis J, Hertzano R, Hof PR, Hollt T, Horwitz GD, Keene CD, Kharchenko PV, Ko AL, Lelieveldt BP, Luo C, Mukamel EA, Pinto-Duarte A, Preissl S, Regev A, Ren B, Scheuermann RH, Smith K, Spain WJ, White OR, Koch C, Hawrylycz M, Tasic B, Macosko EZ, McCarroll SA, Ting JT, Zeng H, Zhang K, Feng G, Ecker JR, Linnarsson S and Lein ES. Comparative cellular analysis of motor cortex in human, marmoset and mouse. Nature. 2021;598:111–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Tzahor E and Dimmeler S. A coalition to heal-the impact of the cardiac microenvironment. Science. 2022;377:eabm4443. [DOI] [PubMed] [Google Scholar]
  • 201.Kim B, Kim H, Kim S and Hwang YR. A brief review of non-invasive brain imaging technologies and the near-infrared optical bioimaging. Appl Microsc. 2021;51:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Brunet J, Walsh CL, Wagner WL, Bellier A, Werlein C, Marussi S, Jonigk DD, Verleden SE, Ackermann M, Lee PD and Tafforeau P. Preparation of large biological samples for high-resolution, hierarchical, synchrotron phase-contrast tomography with multimodal imaging compatibility. Nature protocols. 2023;18:1441–1461. [DOI] [PubMed] [Google Scholar]
  • 203.Sun T, Li Y, Forstera B, Stanic K, Lu S, Steffens S, Yin C, Erturk A, Megens RTA, Weber C, Habenicht A and Mohanta SK. Tissue Clearing Approaches in Atherosclerosis. Methods Mol Biol. 2022;2419:747–763. [DOI] [PubMed] [Google Scholar]
  • 204.Mimitou EP, Lareau CA, Chen KY, Zorzetto-Fernandes AL, Hao Y, Takeshima Y, Luo W, Huang TS, Yeung BZ, Papalexi E, Thakore PI, Kibayashi T, Wing JB, Hata M, Satija R, Nazor KL, Sakaguchi S, Ludwig LS, Sankaran VG, Regev A and Smibert P. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nature biotechnology. 2021;39:1246–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Chang RB. Optogenetic Control of the Peripheral Nervous System. Cold Spring Harb Perspect Med. 2019;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Deisseroth K Optogenetics: 10 years of microbial opsins in neuroscience. Nature neuroscience. 2015;18:1213–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Roth BL. DREADDs for Neuroscientists. Neuron. 2016;89:683–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Claes M, De Groef L and Moons L. The DREADDful Hurdles and Opportunities of the Chronic Chemogenetic Toolbox. Cells. 2022;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Carnevale L, Pallante F, Perrotta M, Iodice D, Perrotta S, Fardella S, Mastroiacovo F, Carnevale D and Lembo G. Celiac Vagus Nerve Stimulation Recapitulates Angiotensin II-Induced Splenic Noradrenergic Activation, Driving Egress of CD8 Effector Cells. Cell Rep. 2020;33:108494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Ross R and Glomset JA. The pathogenesis of atherosclerosis (first of two parts). The New England journal of medicine. 1976;295:369–77. [DOI] [PubMed] [Google Scholar]
  • 211.Tabas I and Glass CK. Anti-inflammatory therapy in chronic disease: challenges and opportunities. Science. 2013;339:166–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Grune J, Yamazoe M and Nahrendorf M. Electroimmunology and cardiac arrhythmia. Nat Rev Cardiol. 2021;18:547–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Schloss MJ, Swirski FK and Nahrendorf M. Modifiable Cardiovascular Risk, Hematopoiesis, and Innate Immunity. Circulation research. 2020;126:1242–1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Ben-Shaanan TL, Azulay-Debby H, Dubovik T, Starosvetsky E, Korin B, Schiller M, Green NL, Admon Y, Hakim F, Shen-Orr SS and Rolls A. Activation of the reward system boosts innate and adaptive immunity. Nature medicine. 2016;22:940–4. [DOI] [PubMed] [Google Scholar]

RESOURCES