Wang SY, Liu X, Li ZM, Deng CX, Chen KR, Zhuang SY, Xu B, Xu TC. Peripheral nerve-mediated glucose lowering: Mechanisms, translational strategies, and future perspectives. World J Diabetes 2026; 17(1): 114535 [DOI: 10.4239/wjd.v17.i1.114535]
Corresponding Author of This Article
Tian-Cheng Xu, MD, PhD, Professor, Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, Jiangsu Province, China. xtc@njucm.edu.cn
Research Domain of This Article
Endocrinology & Metabolism
Article-Type of This Article
Minireviews
Open-Access Policy of This Article
This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Shuai-Yan Wang, Xin Liu, Zi-Mu Li, Chen-Xi Deng, Ke-Ran Chen, Si-Yu Zhuang, Bin Xu, Tian-Cheng Xu, Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
Co-corresponding authors: Bin Xu and Tian-Cheng Xu.
Author contributions: Wang SY and Liu X contribute equally to this study as co-first authors; Xu B and Xu TC contribute equally to this study as co-corresponding authors; Wang SY was responsible for the idea and conceptual framework; Wang SY, Liu X, Li ZM, Deng CX, Chen KR, Zhuang SY wrote the first draft of the manuscript; Xu TC and Xu B reviewed the manuscript and critically revised it for important intellectual content; all authors have reviewed and approved the final version of the manuscript.
Supported by The National Natural Science Foundation, No. 82305376; The Youth Talent Support Project of the China Acupuncture and Moxibustion Association, No. 2024-2026ZGZJXH-QNRC005; The 2024 Jiangsu Province Youth Science and Technology Talent Support Project, No. JSTJ-2024-380; and 2025 Jiangsu Provincial Science and Technology Think Tank Program Project, No. JSKX0125035.
Conflict-of-interest statement: The authors declare that there are no conflicts of interest associated with the publication of this manuscript.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Tian-Cheng Xu, MD, PhD, Professor, Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, Jiangsu Province, China. xtc@njucm.edu.cn
Received: September 23, 2025 Revised: October 23, 2025 Accepted: November 19, 2025 Published online: January 15, 2026 Processing time: 114 Days and 0.1 Hours
Abstract
Glucose homeostasis is crucial for metabolic health, with disruptions leading to hyperglycemia (e.g., diabetes affecting over 589 million adults worldwide in 2025) or hypoglycemia, both associated with severe complications like nephropathy, neuropathy, and cardiovascular disease. The peripheral nervous system (PNS) is a less studied regulator of glucose balance through neural circuits involving the sympathetic, parasympathetic, sensory, and somatic nerves, which interact with organs like the liver, pancreas, and gut. This review aims to synthesize recent advances in PNS-mediated glucose lowering, covering mechanistic foundations, organ-specific pathways, translational interventions (e.g., device-based neuromodulation, pharmacological approaches, closed-loop systems, and regenerative strategies), preclinical/clinical evidence, translational challenges, and future perspectives to provide a roadmap for developing novel, drug-free therapies for dysglycemia.
Core Tip: The peripheral nervous system is an underappreciated regulator of glucose balance. Targeted neuromodulation, like vagus nerve stimulation or focused ultrasound, modulates neural circuits to control liver glucose output, pancreatic hormones, and tissue uptake. Preclinical data show strong metabolic gains; early human trials prove safety. Tackling issues in precision, patient differences, and nerve damage could enable scalable, drug-free treatments for diabetes.
Citation: Wang SY, Liu X, Li ZM, Deng CX, Chen KR, Zhuang SY, Xu B, Xu TC. Peripheral nerve-mediated glucose lowering: Mechanisms, translational strategies, and future perspectives. World J Diabetes 2026; 17(1): 114535
Glucose homeostasis is a tightly regulated process essential for maintaining metabolic health. Disruption of this balance, manifesting as hyperglycemia or hypoglycemia, is closely associated with significant morbidity and mortality. According to the latest estimates by the International Diabetes Federation (2025), over 589 million adults worldwide are living with diabetes, and an additional 1.1 billion of the global population present with prediabetic states such as impaired fasting glucose or impaired glucose tolerance[1]. Persistent hyperglycemia imposes a substantial burden through microvascular and macrovascular complications, including nephropathy, neuropathy, retinopathy, and cardiovascular disease. Conversely, hypoglycemia—although less common—remains a serious, potentially lifethreatening complication, especially in insulintreated individuals, elderly patients, and those in critical care. Severe episodes may result in seizures, cognitive dysfunction, cardiac arrhythmias, and, in extreme cases, death. Importantly, recurrent hypoglycemia can impair counterregulatory hormone responses and diminish autonomic warning symptoms, leading to hypoglycemia unawareness. This condition markedly increases the risk of delayed recognition and treatment of low glucose levels, thereby heightening the likelihood of severe outcomes. These dual threats highlight that effective glucose regulation strategies must not only address hyperglycemia but also prioritize prevention of both symptomatic and unrecognized hypoglycemia[2].
Significant strides have been made in recent years in elucidating the mechanisms by which the peripheral nervous system (PNS) modulates glucose homeostasis, highlighting its multifaceted role in maintaining glycemic stability. Vagus nerve stimulation (VNS) has emerged as a promising approach to enhance insulin sensitivity. VNS exerts its effects through a combination of peripheral and central mechanisms: Efferent cholinergic signaling directly targets pancreatic β-cells and α-cells, promoting insulin secretion while suppressing glucagon release, thereby facilitating glucose uptake and utilization in peripheral tissues; afferent signals from the vagus nerve converge in brainstem nuclei, including the nucleus tractus solitarius (NTS) and dorsal motor nucleus of the vagus (DMV), where central integration modulates autonomic outflow to metabolic organs, coordinating systemic glucose regulation[3]. The sympathetic nervous system (SNS) also exerts a profound influence on glucose regulation, particularly in the context of hepatic gluconeogenesis. Selective inhibition of sympathetic nerve activity has been shown to significantly reduce hepatic gluconeogenesis, leading to a decrease in blood glucose levels. This mechanism primarily involves the attenuation of norepinephrine (NE) release, a neurotransmitter that typically stimulates glycogenolysis and gluconeogenesis in the liver[4]. Peripheral sensory nerves, such as TRPV1 and Piezo2, have garnered attention for their role in detecting nutrients in the gut. These sensory nerves transmit signals to the central nervous system (CNS) via the solitary nucleus in the brainstem, thereby modulating insulin and glucagon secretion to maintain postprandial glucose homeostasis. This sensory feedback loop is crucial for the fine-tuning of glucose levels following nutrient intake. Moreover, the vagus nerve is intricately involved in complex signaling pathways with multiple organs, including the liver, pancreas, and gut, to achieve synchronized metabolic regulation. For instance, the vagus nerve regulates glycogen synthesis in the liver and insulin secretion in the pancreas, facilitating coordinated metabolic responses across organs. Additionally, the role of the SNS in brown adipose tissue (BAT) has been explored, with its activation promoting glucose oxidation to maintain energy balance[5]. Recent advances in PNS-targeted neuromodulation offer novel avenues for glucose regulation. Transcutaneous auricular VNS (taVNS) modulates autonomic tone and peripheral organ function, potentially supporting glycemic control[6]. High-frequency spinal cord stimulation (SCS), though developed for neuropathic pain, can alter sympathetic output and thereby influence hepatic glucose production and peripheral uptake[7]. Optogenetic stimulation of pancreatic β-cells or GLP-1-producing enteroendocrine cells (EECs) has improved insulin secretion and glucose tolerance in preclinical models. Non-invasive focused ultrasound can target vagal or sympathetic pathways without implants[8], and miniaturized wireless microstimulators offer long-term, closed-loop autonomic modulation[9]. These approaches enable precise manipulation of metabolic neural circuits and pave the way for integrated neuro-metabolic control systems.
This review synthesizes recent progress in peripheral nerve-mediated glucose lowering, covering mechanistic foundations, organ-specific neural pathways, and translational interventions (Figure 1). We first outline the physiological and anatomical basis of PNS-mediated glucose regulation, followed by an analysis of well-characterized neural control mechanisms. We then examine current and emerging translational strategies, encompassing device-based neuromodulation, pharmacological interventions, integrated closed-loop systems, and regenerative approaches, and conclude with translational challenges and future perspectives. Our aim is to provide a roadmap for advancing PNS-targeted modulation as a novel therapeutic approach for dysglycemia.
Figure 1 Peripheral nerve-targeted neuromodulation for glucose control.
The peripheral nervous system regulates multiple metabolic organs, including the liver, adipose tissue, pancreas, skeletal muscle, and gut, to maintain glucose homeostasis. Key actions include inhibiting hepatic gluconeogenesis and enhancing glycogen synthesis; modulating lipolysis and brown adipose tissue thermogenesis; regulating insulin secretion from β-cells and glucagon release from α-cells; promoting glucose uptake and utilization in skeletal muscle; and enhancing gut hormone (e.g., GLP-1) secretion to improve glucose metabolism. This mechanism provides potential therapeutic targets for neuromodulation in metabolic disorders such as diabetes. BAT: Brown adipose tissue. Created in BioRender (Supplementary material).
PHYSIOLOGICAL AND ANATOMICAL BASIS OF PNS IN GLUCOSE REGULATION
Overview of PNS in metabolic homeostasis
Traditionally, the role of the autonomic nervous system (ANS) in metabolism has often been oversimplified as a dichotomy: The sympathetic nervous system (SNS) mediates fight or flight responses that promote energy mobilization, while the parasympathetic nervous system (PSNS; specifically the vagus nerve) supports rest and digest functions that facilitate energy storage[10]. However, cutting-edge research has fundamentally transformed this perspective, revealing that the PNS functions as a sophisticated network capable of refined, dynamic, and organ-specific metabolic regulation. Far from a simple on/off switch, it constitutes a central command system that continuously senses, integrates, and responds to changes in the internal and external environment to maintain whole-body energy homeostasis[11].
This regulatory network comprises four major components, each playing a distinct yet highly coordinated role in blood glucose regulation. SNS generally exerts antagonistic effects by elevating blood glucose through multiple mechanisms: Stimulating hepatic glycogenolysis and gluconeogenesis (thus increasing hepatic glucose output), suppressing insulin secretion (pancreas), promoting glucagon release, enhancing lipolysis (adipose tissue), and restricting glucose utilization in skeletal muscle[12]. PSNS primarily mediated by the vagus nerve, and it acts to lower blood glucose in the postprandial state through multiple integrated mechanisms: Stimulating the secretion of hormones such as insulin and GLP-1 from the pancreas, enhancing hepatic glycogen synthesis, and modulating gastrointestinal motility and nutrient absorption.
Acting as metabolic sensors, sensory nerves are distributed throughout organs such as the liver, pancreas, gastrointestinal tract, and adipose tissue. They express various receptors (e.g., TRPV1, Piezo2) that detect signals including glucose, free fatty acids, hormones (e.g., insulin, leptin), and mechanical stress. This information is relayed to the CNS via spinal or vagal afferent pathways. Dysfunction in this sensory signaling is a key element in complications such as diabetic peripheral neuropathy and represents an important target for neuromodulation therapies[13].
Although somatic nerves do not directly innervate internal organs, somatic nerves function as the controllers of skeletal muscle contraction—effectively regulating the body’s largest glucose-consuming tissue and thereby exerting substantial influence over glucose disposal.
The contemporary view characterizes peripheral neural regulation of metabolism as a multimodal, bidirectional communication system. This system not only executes central commands but also forms closed-loop feedback circuits via sensory afferents. Furthermore, it engages in complex interactions such as immune-neural crosstalk (e.g., neuromodulation of macrophage function) and gut microbiota-brain axes, enabling precise coordination of glucose homeostasis, energy allocation, nutrient digestion and absorption, and immunometabolic coupling[14].
Organ-specific neural pathways
Regulation of blood glucose by the PNS is achieved through direct innervation of key metabolic organs. Table 1 systematically summarizes the neural innervation, physiological functions, and underlying molecular mechanisms in major organs.
Table 1 Innervation of key metabolic organs and their roles in blood glucose regulation.
Organ
Nerve type
Innervation description
Role in glucose regulation
Key neurotransmitters/pathways
Liver
SNS
Predominantly innervated by the greater splanchnic nerve
Primarily releases norepinephrine, which acts on α- and β-adrenergic receptors
PSNS
Innervated by the hepatic branch of the vagus nerve
Activation promotes glycogen synthesis and suppresses gluconeogenesis, leading to reduced hepatic glucose output
Releases acetylcholine, acting primarily on M3 muscarinic receptors
Sensory nerves
Express various metabosensors
Detect intrahepatic signals such as glucose levels, ATP/AMP ratio, and inflammatory cytokines, and relay this information to the brainstem and hypothalamus
/
Pancreas
SNS
Originates from the celiac ganglion
Activation inhibits insulin secretion from β-cells while stimulating glucagon release from α-cells
Releases norepinephrine, which acts on α2-adrenergic receptors to suppress insulin secretion
PSNS
Derived from the pancreatic branch of the vagus nerve
Activation (particularly postprandially) strongly stimulates the secretion of both insulin and glucagon, exhibiting a biphasic effect
Primarily releases acetylcholine acting on M3 receptors, promoting insulin secretion via the IP3/PKC signaling pathway. Additionally, neuropeptides such as VIP and PACAP are involved in enhancing secretory responses
Sensory nerves
Densely distributed throughout the islets of Langerhans and surrounding pancreatic tissue
Detect local insulin and glucose levels, and participate in the feedback regulation of pancreatic islet function
/
Adipose tissue
SNS
Heavily innervates both white and brown adipose tissue
Activation stimulates lipolysis, increasing the release of FFAs, which may indirectly affect hepatic glucose output and muscle glucose utilization via lipotoxicity
Releases norepinephrine, which primarily acts on β3-adrenergic receptors to promote lipolysis
Sensory nerves
Provide feedback on adipose tissue metabolic status
Detect levels of adipokines such as leptin and adiponectin, and relay energy storage signals to the central nervous system
Express receptors such as LepR and TrkB, the latter being a high-affinity receptor for BDNF
Releases norepinephrine, which acts on α1-adrenergic receptors (causing vasoconstriction) and β2-adrenergic receptors (promoting vasodilation and enhancing glucose uptake). Key mechanisms in contraction-induced
Somatic motor nerves
Regulate voluntary muscle contraction
Muscle contraction per se serves as the most potent stimulus for glucose uptake and utilization, primarily through AMPK activation and enhanced GLUT4 translocation
/
Gastrointestinal tract
PSNS
Vagus nerve (afferent/efferent) SNS
Enhances intestinal motility, stimulates secretion, and increases nutrient absorption surface area, thereby indirectly modulating the rate of blood glucose elevation
/
Sensory nerves
Extremely abundant
Play an essential role. They detect nutrients such as glucose, fatty acids, and amino acids, as well as hormones (e.g., GLP-1, PYY, CCK), and transmit these signals via vagal afferents to the NTS. This triggers gut-brain axis reflexes that preemptively regulate insulin secretion (cephalic phase insulin release) and promote satiety
Express a wide range of nutrient-sensing receptors, including but not limited to GLP-1R, CCKAR, SGLT1, and GPR40
The organ-specific neural innervation described above does not operate in isolation but is integrated within a sophisticated closed-loop reflex arc, which is central to understanding peripheral nervous glycemic control. This classic reflex arc consists of four essential components, which in the context of metabolic regulation are manifested as follows: Peripheral nerves act as vigilant detectors scattered across key organs like the portal vein, liver, gut, and pancreas. These endings serve as chemical and mechanical sensors, constantly tracking elements such as blood sugar, available nutrients, hormone levels, and various metabolites[15]. In the incoming signal route, these sensory inputs are converted by neurons into electrical impulses, transmitting signals through two principal pathways: Vagal afferents, which convey visceral information from thoracoabdominal organs to the brainstem, and spinal afferents, which relay somatic and visceral input via dorsal root ganglia. Vagal afferents constitute the parasympathetic sensory component, whereas spinal afferents are anatomically distinct and independent of parasympathetic circuits. Collectively, they channel metabolic information to the brain. For vagal paths, the neuron bodies are located in the nodose ganglion, with their extensions connecting to the NTS in the brainstem[16]. Acting as the main hub for processing, the NTS gathers and begins to merge these internal signals. From there, data flows to advanced brain areas like the hypothalamus, including spots such as the arcuate, paraventricular, and ventromedial nuclei that oversee energy and hormone systems, the parabrachial nucleus, and even the cerebral cortex, which handles feelings like appetite. Collectively, these form a core command center that analyzes the inputs thoroughly and crafts suitable outgoing instructions[17].
These coordinated directives are sent out through sympathetic channels starting in areas like the hypothalamus, moving to the spinal cord's intermediolateral column, then to follow-up neurons in sympathetic clusters, and finally to the intended organs. Similarly, parasympathetic routes begin at the dorsal vagal complex, extend through preganglionic vagal fibers to local postganglionic neurons, and reach their destinations. Together, they fine-tune processes like insulin release, liver sugar production, muscle sugar absorption, and digestive operations, bringing blood glucose back to its ideal level[18] (Table 2).
Table 2 Overview of the neural regulation of glucose metabolism.
Component
Description
Key structures/examples
Primary function
Ref.
Sensors
Sensory nerve endings acting as chemoreceptors and mechanoreceptors
Portal vein, liver, intestine, pancreas
Monitor blood glucose, nutrients, hormones, and metabolites
This closed-loop system explains why blood glucose regulation is both rapid and precise. For example, postprandial nutrient sensing in the gut is transmitted via vagal afferent nerves to the CNS, which can then—even before blood glucose rises significantly—trigger cephalic phase insulin release through vagal efferent pathways, thereby preparing the body for the impending glucose load. Similarly, at the onset of exercise, sensory signals from muscles are relayed to the CNS, which modulates sympathetic output to the liver and pancreas to ensure adequate glucose supply to active muscles.
The PNS is far more than a passive set of wires; it constitutes an active and intelligent regulatory network. This integrated physiological perspective highlights the foundational role of sensory information and central integration in maintaining glucose homeostasis. Furthermore, it provides a physiological basis for integrating such mechanisms with artificial intelligence (AI) and continuous glucose monitoring (CGM) technologies to develop intelligent, real-time feedback control systems[19].
WELL-CHARACTERIZED MECHANISMS OF PNS-ENDOCRINE-IMMUNE REGULATION
Energy metabolic homeostasis relies on the intricate interplay among the nervous, endocrine, and immune systems. Traditional perspectives have predominantly focused on isolated autonomic neural control or endocrine regulation. However, contemporary studies have demonstrated that these mechanisms are interdependent—central neural circuits integrate neural and endocrine signals to coordinate immune responses, thereby fine-tuning blood glucose levels within a dynamic equilibrium[20]. Within the PNS, both the PSNS and SNSs play pivotal roles in glucose homeostasis. These divisions establish tight functional coupling with the CNS through positive and negative feedback loops, collectively contributing significantly to the reduction of blood glucose levels.
Vagal parasympathetic pathways
The PSNS, particularly the vagus nerve, is critically involved in anabolic processes and plays an essential role in energy storage. Efferent fibers of the vagus nerve originating from the DMV project to pancreatic β-cells, where the released acetylcholine (ACh) activates M3 muscarinic receptors, triggers calcium influx, and enhances glucose-stimulated insulin secretion (GSIS)[21]. In addition to its insulinotropic effect, vagal signaling also suppresses glucagon secretion from pancreatic α-cells, thereby exerting a dual glucose-lowering effect. Furthermore, subsequent studies have demonstrated that vagal activity promotes hepatic glycogen synthesis and suppresses gluconeogenesis via both neural and humoral pathways: Insulin acts on hypothalamic AgRP neurons to activate vagal efferents, which project to the liver and stimulate resident macrophages, such as Kupffer cells, to locally release interleukin-6. This, in turn, downregulates the expression of gluconeogenic enzymes such as PEPCK, thereby reducing hepatic glucose output. Notably, the regulation of glycemia by the vagus nerve is not solely dependent on ACh. Evidence suggests that neuropeptides, such as vasoactive intestinal peptide, act as co-transmitters and modulate glucose metabolism through endothelial insulin signaling, highlighting the mechanistic diversity of vagal-mediated metabolic control[22]. In summary, the vagal parasympathetic pathway regulates systemic metabolism by enhancing insulin secretion, suppressing hepatic glucose production, and promoting glycogen storage.
Sympathetic pathways
In contrast to the PSNS, the SNS primarily exerts catabolic actions, lowering blood glucose by increasing energy expenditure. In the liver, sympathetic innervation stimulates glycogenolysis and gluconeogenesis via α1- and β2-adrenergic receptors, which elevate intracellular cAMP levels, activate protein kinase A, and subsequently phosphorylate glycogen phosphorylase. This mechanism is typically engaged during fasting or stress to increase circulating glucose levels in order to meet heightened energy demands. Conversely, suppression of hepatic sympathetic tone—achieved through pharmacological or genetic interventions—can inhibit gluconeogenic pathways and improve insulin sensitivity.
Notably, SNS activation also promotes glucose oxidation through the activation of BAT. Sympathetic fibers innervating BAT release NE, which binds to β3-adrenergic receptors and triggers thermogenesis mediated by UCP1, thereby consuming substantial amounts of glucose and fatty acids[23]. This process is independent of insulin and represents a unique physiological function of the SNS, particularly prominent during cold exposure or administration of adrenergic agonists.
Recent studies have further revealed that the sympathetic axis can directly modulate immune cell function. NE, acting via β2-adrenergic receptors, promotes the polarization of anti-inflammatory M2 macrophages in adipose tissue, thereby suppressing pro-inflammatory cytokine expression and improving insulin sensitivity. For example, Lu et al[24] demonstrated that electroacupuncture suppressed sympathetic activity in epididymal white adipose tissue of mice, markedly reducing neuropeptide Y (NPY) release and alleviating adipose tissue inflammation. However, chronic SNS activation may lead to catecholamine resistance and sympathetic neuropathy, thereby exacerbating metabolic dysfunction. Together, these findings underscore the importance of SNS-immune cell interactions in metabolic regulation. Therefore, the SNS modulates glucose metabolism through two interconnected mechanisms: Directly regulating hepatic glucose production and BAT-mediated glucose disposal, as well as integrating immune-metabolic pathways to influence tissue insulin sensitivity.
Sensory afferent pathways
Afferent nerves convey peripheral metabolic information to the CNS, enabling coordinated regulation of glucose homeostasis through multi-level, bidirectional communication between the PNS and CNS. Within the CNS, the hypothalamic paraventricular nucleus and arcuate nucleus are considered key therapeutic targets for metabolic disorders. Due to their unique structural features, these nuclei can sense and receive nutrient and hormonal signals from peripheral organs such as the liver, adipose tissue, and skeletal muscle, and relay metabolic signals via the ANS.
First, sensory afferent nerves can significantly influence glucose and insulin delivery and exchange efficiency by modulating local tissue blood flow. Studies have shown that gastrointestinal peptidergic sensory neurons release calcitonin gene-related peptide and substance P, inducing vasodilation and thereby enhancing nutrient and hormone exchange. Concurrently, the hypothalamic median eminence dynamically regulates vascular permeability, enabling real-time sensing of circulating metabolites or inflammatory signals, and mediates neuroendocrine output to precisely control systemic glucose homeostasis[25].
Furthermore, recent findings have identified the TRPV1—a molecular sensor expressed on sensory neurons activated by capsaicin—as a potential therapeutic target for improving insulin resistance. TRPV1 activation enhances glucose oxidation and elevates intracellular ATP levels via CAMKK2 and AMPK activation. However, experimental data regarding the effect of capsaicin on intracellular signaling molecules involved in glucose uptake remain limited, and the underlying mechanisms are yet to be fully elucidated[26].
In addition, in the gastrointestinal tract, EECs function as chemoreceptors, releasing peptides such as CCK and GLP-1. These peptides act on vagal afferent fibers, transmitting signals to the NTS in the brainstem, where visceral feedback is integrated with hypothalamic and limbic system inputs to enhance glucose tolerance[27]. Notably, the gut-brain-liver axis operates as a closed-loop feedback system that embodies multi-organ reflex integration: Following nutrient absorption (e.g., glucose and lipids), EECs secrete GLP-1 and CCK, which not only act via endocrine pathways but also activate vagal afferent terminals innervating the gut. Signals are relayed to the NTS and subsequently transmitted via vagal efferents to the liver, where they suppress the expression of key gluconeogenic genes. The resulting reduction in hepatic glucose output lowers blood glucose levels, which in turn attenuates stimulation of intestinal EECs, thereby completing a physiological feedback loop[28].
In summary, sensory afferent pathways represent critical channels of metabolic communication, linking multi-organ function with central regulatory hubs through TRPV1-dependent signaling, chemosensory mechanisms, and vascular control, ultimately ensuring effective glycemic regulation.
TRANSLATIONAL APPROACHES FOR PNS-MEDIATED GLUCOSE MODULATION
Building upon the established roles of peripheral nerves in glucose homeostasis, significant efforts are underway to translate these mechanistic insights into clinically viable therapeutic strategies. This section delineates the major translational approaches, categorized by their underlying technological or biological principles, with a specific focus on their distinct characteristics in modulating glucose metabolism.
Unlike simple hypoglycemic drugs, device-based neuromodulation technology enables accurate and reversible interventions for glucose regulation. For example, VNS uses a subcutaneously implanted pulse generator and two leads (placed around the anterior vagal trunk near the gastroesophageal junction) to deliver signals inducing intermittent vagal blockage. Follow-up studies have shown that this therapy is effective in improving patients' lipid profiles, glycated hemoglobin (HbA1c) levels, systolic and diastolic blood pressure, as well as their quality of life[29]. Its key advantages include immediate reversibility and adjustable parameters, enabling real-time optimization. Preclinical studies demonstrate VNS reduces hyperglycemia in diabetic rodent models by modulating autonomic outflow[30].
As a non-invasive alternative, taVNS stimulates auricular vagus nerve branches. Systematic reviews confirm its safety profile, with minimal adverse events across clinical applications[31]. SCS targets dorsal spinal structures. Rodent studies demonstrate kilohertz-frequency SCS differentially modulates excitatory and inhibitory neuronal activity in the dorsal horn, suggesting potential for fine-tuning autonomic outflow[32].
Pharmacological neuromodulation: Systemic action
Pharmacological neuromodulation employs chemical agents to modulate peripheral neural activity for glucose regulation, characterized by well-defined stimulatory effects yet limited specificity due to systemic actions. Cholinergic agonists, such as carbachol, enhance GSIS by activating pancreatic M3 receptors, amplifying the first-phase insulin response. In high-fat diet-induced insulin-resistant mice, carbachol (15-50 nmol/kg) normalized glucose tolerance by increasing insulin secretion efficiency (392%-479% potentiation vs controls), though its effects were not islet-specific, as ileal contractility remained unchanged[33]. β-adrenergic blockers like metoprolol reduce hepatic glucose production via β2-receptor antagonism. At high doses (≥ 100 mg/day), metoprolol significantly suppressed terbutaline-induced glucose output [area under the curve (AUC) reduction: 2424-2437 mg/dL·minute] in heart failure patients, whereas carvedilol showed no effect, highlighting receptor-subtype-dependent specificity limitations[34]. TRPV1 agonists (e.g., capsaicin) mitigate hyperglycemia-induced liver injury by upregulating mitochondrial fusion protein OPA1, reducing reactive oxygen species and apoptosis in diabetic mice. However, TRPV1’s broad expression in sensory neurons and hepatocytes results in off-target effects, restricting tissue-specific modulation[35]. These agents demonstrate clear dose-responsive efficacy but face challenges in target selectivity.
Integrated closed-loop systems: Dynamic control
Integrated closed-loop systems enable real-time glucose monitoring and adaptive neuromodulation by combining continuous glucose sensors, control algorithms, and implantable stimulators. These systems dynamically adjust neural stimulation parameters in response to glycemic fluctuations, exemplifying the core feature of real-time glucose sensing and immediate stimulation adjustment[36]. For instance, a rat study demonstrated a self-powered VNS device that modulated gastric peristalsis based on real-time motility measurements, achieving a 38% reduction in body weight through adaptive stimulation[37]. Similarly, in a murine model of autoimmune diabetes, closed-loop pancreatic nerve stimulation (10 Hz, 450 μA) suppressed pro-inflammatory cytokines and autoreactive T-cell proliferation, effectively halting disease progression without adverse effects[38]. Commercial hybrid closed-loop systems, though primarily insulin-delivery focused, validate the translational potential of this architecture: They integrate CGM data with algorithms to autonomously titrate therapy, improving time-in-range (TIR) by 10%-15% in clinical trials[39]. Such systems underscore the feasibility of extending closed-loop principles to neuromodulation, where neural signals could similarly drive precise, glucose-responsive stimulation.
Regenerative and genetic approaches: Long-term effects
Regenerative and genetic strategies offer potential long-term glucose modulation through sustained neurotrophic factor enhancement or precise gene editing, yet face significant risks and ethical challenges. Neurotrophic factors like GDNF demonstrate durable protective effects in diabetic enteric neuropathy. In streptozotocin-induced diabetic mice, GDNF overexpression prevented hyperglycemia-induced neuronal apoptosis and loss of nitrergic inhibitory neurons via PI3K/Akt pathway activation, with benefits persisting for weeks[40]. Similarly, BDNF gene therapy in obese mice promoted thermogenesis and improved insulin sensitivity long-term by upregulating UCP1 in adipose tissue. CRISPR-Cas9 editing further enables permanent genomic alterations. Silencing Fabp4 via CRISPR interference in adipocytes reduced body weight by 20% in high-fat-diet mice, with effects maintained over 6 weeks through suppressed inflammation and improved hepatic metabolism[41]. However, CRISPR systems pose substantial risks, including off-target effects (e.g., unintended DNA cleavage) and delivery challenges due to Cas9’s large size. Ethical concerns surrounding germline editing and long-term safety in humans remain unresolved, limiting clinical translation despite promising preclinical efficacy (Table 3).
Table 3 Typical patents for precise regulation of blood glucose based on the peripheral nervous system.
Number
Name
First inventor
Core mechanism
Function
Ref.
US10722714B2
Methods and systems for glucose regulation
Arnold W Thornton
Modulating peripheral nerves affects insulin/GLP-1 secretion to regulate glycemia
A range of preclinical studies has shown that peripheral neuromodulation can beneficially modulate glucose metabolism in both small and large animals. VNS approaches have demonstrated positive effects[42-49]. TaVNS in rodent models lowered fasting glucose, improved glucose tolerance, and favorably modulated hormones such as GLP-1 and ghrelin. Other kinds of VNS, including invasive stimulation of the cervical vagus or direct pancreatic branches, have increased insulin secretion, enhanced pancreatic blood flow, and rapidly reduced blood glucose. Ultrasound stimulation has also demonstrated great potential[50-53]. Hepatic peripheral focused ultrasound stimulation (pFUS) has produced consistent metabolic improvements in rodent and swine models of insulin resistance, including reductions in fasting glucose and insulin, improved Homeostasis Model Assessment of Insulin Resistance (HOMA-IR), enhanced glucose tolerance, and increased glucose infusion rates (GIRs) during hyperinsulinemic-euglycemic clamps (HECs)[50]. Optogenetic activation of pancreatic cholinergic vagal efferents has further confirmed the potential of neuromodulation to acutely elevate insulin levels and improve glycemic control[54-56]. Overall, these findings consistently demonstrate that peripheral neuromodulation can improve glycemic control in animal models, providing a strong rationale for clinical translation.
Clinical studies
Clinical evaluation of peripheral neuromodulation for glycemic control remains limited, with only a few trials exploring its feasibility and metabolic effects in humans. Ashe et al[53] reported in a first-in-human feasibility study, hepatic pFUS was applied to patients with type-2 diabetes for 15 minutes daily over three consecutive days. The intervention was well tolerated and resulted in a statistically significant reduction in fasting insulin and HOMA-IR (P = 0.01). Although fasting glucose remained unchanged, it suggests a modest improvement in insulin sensitivity over a short treatment window. In a larger randomized, sham-controlled study[49], Kufaishi et al[49] evaluated cervical transcutaneous VNS (tVNS) in people with diabetes and autonomic neuropathy and found that, while group-level CGM outcomes were neutral, a predefined type-1 diabetes subgroup experienced a clinically meaningful 11.6% reduction in glucose coefficient of variation (CV) after 8 weeks of active stimulation, demonstrating that neuromodulation can reduce glycemic variability in susceptible patients. SCS, primarily developed for painful diabetic neuropathy, has also been associated with secondary metabolic benefits: In a post-study investigation of a randomized trial of 10 kHz SCS[57], significant and sustained pain relief was accompanied by reductions in HbA1c and body weight in patients with type-2 diabetes and elevated baseline values, although these were secondary endpoints. Collectively, these studies indicate that peripheral neuromodulation is technically feasible and generally safe in human populations.
Bench-to-bedside translation issues
Preclinical models have consistently demonstrated substantial metabolic benefits following peripheral neuromodulation interventions. For instance, in rodents subjected to hepatic pFUS, fasting insulin levels significantly decreased and HOMA-IR improved, while in swine, GIR—a HEC measure of insulin sensitivity—was markedly increased (pFUS group showed higher steady-state GIR and elevated GIR AUC vs controls)[50]. In contrast, the first-in-human pFUS feasibility trial in type 2 diabetes patients, applying 15 minutes of hepatic ultrasound for 3 consecutive days—produced only a modest statistically significant reduction in fasting insulin and HOMA-IR (P = 0.01), with no significant change in fasting glucose[53]. A key contributor to this translation gap lies in the choice of outcome metrics. Animal studies utilize high-sensitivity metabolic assessments, such as oral glucose tolerance test, AUC, HOMA-IR, and GIR—that can detect subtle shifts in glucose handling[51]. Clinical studies, however, often depend on composite or long-term outcomes like CGM metrics (CV, TIR) and HbA1c, which may require longer duration to reflect meaningful changes. For example, taVNS in humans did not improve CGM metrics over 8 weeks overall; only in a type 1 diabetes subgroup did CV decline by 11.6%, P = 0.009: A much smaller effect size relative to preclinical outcomes[49,50]. Stimulation parameters and anatomical targeting also differ dramatically. Preclinical protocols typically use precisely targeted, higher-intensity stimuli, activating specific plexuses such as the hepatic portal system for longer duration[51-53]. By contrast, clinical pFUS is limited to short daily sessions and conservative parameters for safety, likely reducing efficacy. Similarly, taVNS in animal models may use extended stimulation protocols at higher intensity[42-45], whereas human subjects underwent limited daily stimulation (4 times/day for 1 week, then 2 times/day for 8 weeks) with self-administered devices[50]. Moreover, model characteristics differ: Animal models often involve young, otherwise healthy subjects with induced or genetic diabetes of short duration, lacking comorbidities. Human participants, conversely, often have established, long-duration type-2 diabetes complicated by autonomic neuropathy, obesity, and cardiovascular disease—factors likely dampening neuromodulatory efficacy. Finally, differences in neuroanatomy, innervation density, and nerve responsiveness between species may hinder translating rodent-targeted interventions to human contexts. Activation thresholds and distribution of sensory afferents in humans may not mirror those in animal models, making direct parameter translation unreliable. In conclusion, although preclinical studies have fully demonstrated that metabolic regulation can be achieved through peripheral neural control, the clinical benefits in human studies remain limited. This disparity mainly stems from differences in outcome indicators, stimulation parameters, subject characteristics, and neurophysiological mechanisms, etc. Therefore, it is of vital importance to conduct an in-depth analysis of the mechanisms and practical obstacles that cause the gap.
Comparative analysis of neuromodulation modalities
Given the variety of peripheral neuromodulation methods explored for metabolic control, a structured comparative framework was crucial to understand their different mechanisms and potential for translation. Each modality engages different neural targets, leading to heterogeneous biological effects and patient suitability. These approaches can be generally divided based on the type of energy delivered and the level of invasiveness into non-invasive electrical stimulation, invasive electrical stimulation, acoustic/mechanical neuromodulation and optogenetic modulation. Such a systematic taxonomy not only facilitates a mechanistic comparison of each technique’s neurophysiological basis, advantages and limitations, but also offered clear concepts for recognizing proper clinical applications in glucose regulation (Tables 4 and 5).
Table 4 Summary of representative neuromodulation and biophysical stimulation interventions for glycemic regulation in experimental and clinical models.
Intervention methods
Experimental subject
Intervention duration
Stimulus parameter
Main results
Ref.
taVNS
ZDF rats with T2D
4 weeks, 30 minutes daily
2 mA, 15 Hz
The blood glucose level in the taVNS group decreased from the second week and remained below the baseline level throughout the observation period
The fasting blood glucose was decreased (P < 0.05, P < 0.01) and the insulin receptor expression level in the liver, the skeletal muscle and the pancreas was increased (P < 0.05, P < 0.01, P < 0.001)
No significant changes were observed in CGM metrics between treatment arms, while individuals with T1D and DAN decreased their CV after 8 weeks of tVNS treatment
Daily application of pFUS for 3 minutes can maintain the circulating blood glucose of T2D animals to normal levels, increase glucose uptake and glycogen accumulation in peripheral tissues, especially skeletal muscles, and comprehensively improve glucose tolerance and insulin sensitivity
The mice in the T2D-LIPUS group displayed significantly lower area under the curve of glucose tolerance tests and insulin tolerance tests and fasting serum insulin levels compared to the T2D-sham group
The translation of PNS based interventions for glucose regulation from preclinical research to clinical practice faces several substantial challenges. These barriers span scientific, technological, clinical, and ethical domains, each requiring careful consideration to ensure safe and effective application.
Neuroanatomical and functional variability
Significant interspecies differences in neuroanatomy and neural function complicate the extrapolation of animal findings to humans. For instance, vagal innervation patterns to metabolic organs such as the liver and pancreas differ notably between rodents and humans[58]. Rodent models often exhibit more diffuse and accessible neural pathways, whereas human anatomy involves deeper and more variable nerve distributions, which can affect the accuracy and efficacy of neuromodulatory interventions.
Furthermore, considerable interindividual variability in autonomic nerve architecture exists, which is often exacerbated in diabetic patients with underlying neuropathic changes[59]. These discrepancies contribute to inconsistent responses to neuromodulation modalities such as taVNS when moving from standardized animal models to heterogeneous human populations. Personalized mapping of nerve anatomy and function may be necessary to optimize stimulation sites and parameters.
Impact of diabetic neuropathy
The presence of diabetic peripheral and autonomic neuropathy may severely compromise the efficacy of PNS-targeted therapies. Structural and functional impairments, including axonal degeneration and demyelination, can diminish electrical conduction efficiency and alter neurotransmitter release[60]. This neurodegeneration not only reduces the responsiveness to external stimulation but also disrupts endogenous regulatory mechanisms.
Additionally, neuropathy-induced changes in receptor expression and signaling pathways may further distort metabolic responses to neural stimulation[61]. For example, altered adrenergic receptor sensitivity in diabetic patients can lead to aberrant sympathetic outflow, undermining the glucose-lowering effects of sympathetic modulation. Therefore, patient stratification based on detailed neurological assessments is essential to identify suitable candidates for neuromodulatory treatments.
Device-related complications and long-term adaptations
Although implantable neuromodulation devices (e.g., VNS and SCS) are established in neurology and pain management, their long-term safety and efficacy in metabolic disorders remain uncertain. Risks include surgical complications, infection, fibrotic encapsulation, electrode migration, and hardware malfunction. These issues are particularly relevant in diabetic patients, who often exhibit impaired wound healing and increased infection risk.
Moreover, chronic electrical stimulation may induce neural adaptation or tolerance, leading to attenuated therapeutic effects over time[62]. This phenomenon, often referred to as “stimulation fatigue”, necessitates the development of dynamic stimulation paradigms that can adapt to changing neural plasticity. Improving device biocompatibility, refining electrode design, and optimizing stimulation paradigms are active areas of investigation to enhance long-term efficacy.
Lack of stimulation specificity and off-target effects
Achieving targeted neuromodulation without activating unintended pathways remains a significant hurdle. Non-invasive approaches such as taVNS often lack spatial precision, potentially stimulating cardiac or respiratory branches of the vagus nerve and causing adverse effects[63]. These off-target effects not only limit therapeutic windows but also raise safety concerns in vulnerable populations.
Similarly, pharmacological agents like beta-blockers may modulate sympathetic activity but carry cardiovascular risks that limit their use in diabetic populations. Enhancing the specificity of both device-based and pharmacological interventions through advanced targeting technologies or combination therapies is critical for future success.
Regulatory and ethical hurdles
The regulatory pathway for neuromodulation therapies is still evolving. Agencies such as the Food and Drug Administration require robust long-term efficacy and safety data, particularly for implantable systems. The lack of standardized endpoints for metabolic neuromodulation further complicates trial design and regulatory approval. Demonstrating not only glycemic improvement but also reduced morbidity and mortality will be essential for widespread adoption.
Ethical considerations including informed consent in vulnerable populations (e.g., elderly patients with cognitive impairment), data privacy in closed-loop systems, and equitable access to high-cost therapies also warrant serious attention. The potential for neuromodulation to exacerbate health disparities must be addressed through inclusive trial designs and accessible technology platforms.
The translation of PNS-mediated glucose modulation strategies is fraught with multifaceted challenges, ranging from biological variability and neuropathic complications to technical and ethical limitations. Addressing these issues will require interdisciplinary collaboration to develop personalized stimulation protocols, improve device technology, and establish clear regulatory frameworks. Overcoming these barriers is essential to realize the full clinical potential of neuromodulation in diabetes care.
FUTURE PERSPECTIVES
As reviewed in previous sections, PNS is emerging as a promising model for treating diseases such as obesity, diabetes, and other diseases caused by glucose homeostasis disorders, yet its clinical translation requires overcoming current anatomical, technical, and accessibility barriers. Future development must shift towards multi-target, adaptive and personalized solutions. Here we outline the following key directions.
Multitarget synergistic stimulation
Current PNS approaches predominantly target single pathways (e.g., vagal hepatic branches or sciatic nerves), failing to harness the full complexity of neural-glucose crosstalk. Future advances should enable synergistic stimulation for multiple targets. For instance, joint regulation of vagal pancreatic branches (enhancing insulin secretion via M3 receptors) and sympathetic BAT-activating fibers (promoting glucose uptake via UCP1-mediated thermogenesis) could amplify glycemic control beyond monotherapies[64]. Developing multi-electrode arrays capable of selectively activating efferent/afferent fibers across vagal, sympathetic, and somatic nerves will be critical to replicate these physiological networks.
Personalized neuromodulation based on neural phenotypes
Anatomical variability (e.g., 45% of auricular regions innervated by non-vagal nerves) and physiological heterogeneity (e.g., autonomic neuropathy in long-term diabetes) necessitate patient-specific neuromodulation[65]. Future systems must integrate neural phenotyping to tailor stimulation parameters. For example, tVNS efficacy could be optimized by identifying patient-specific targets in the auricle using functional magnetic resonance imaging (MRI)-guided targeting[66], while avoiding regions prone to off-target effects (e.g., laryngeal activation). Genetic profiling may further stratify patients: Those with NPY-receptor polymorphisms might benefit from adjusted sympathetic stimulation protocols to counteract NPY-mediated hepatic gluconeogenesis. This precision approach could resolve the U-shaped efficacy curve dilemma, where standardized parameters yield suboptimal outcomes[67].
For the clinical translation to be possible, it was essential to create a reproducible format for stimulation standardization. High resolution ultrasound along with functional MRI imaging and electrophysiological profiling could potentially define anatomical landmarks and functional thresholds of neural activation which would enable a reference format to guide parameter selection among patients; meanwhile, physiological biomarkers like heart rate variability, circulating GLP-1 and skin conductance were able to offer continuous and objective measures of autonomic and metabolic states which allowed for real time adjustment of stimulation intensity and frequency. Multimodal datasets, when combined with algorithms powered by AI, had the potential to dynamically adjust stimulation parameters according to an individual's physiological state while keeping within the limits for safety. This combined method of standardized personalization did not just account for differences between individuals but also created a reproducible path towards scalable neuromodulation protocols.
AI-driven closed-loop optimization
Real-time adaptation to dynamic metabolic demands is unattainable with open-loop devices. Closed-loop systems integrating CGM, neural activity sensors (e.g., vagal afferent traffic), and AI algorithms will enable autonomous measurement and control. Machine learning models trained on multimodal data (glucose trends, heart rate variability, inflammatory markers) could predict optimal stimulation parameters. For instance, reinforcement learning algorithms might adjust VNS frequency (5-30 Hz) based on Metabolic biomarkers and neurophysiological recordings (like real-time GLP-1 secretion and hepatic glucose output), mimicking physiological feedback. Crucially, such systems must incorporate safety constraints (e.g., capping stimulation intensity to prevent laryngeal spasm) and address latency challenges via edge computing. Early prototypes combining CGM with tVNS reduced HbA1c variability by 60% in pilot trials, underscoring the feasibility of adaptive control[68].
Global accessibility design
To ensure equitable adoption, PNS technologies must transcend cost and infrastructure barriers. Modular, low-power devices using smartphone-based controllers could democratize access. For example, simplified tVNS units with reusable electrodes and Bluetooth connectivity could be deployed in resource-limited settings, leveraging telemedicine for parameter calibration[69]. Additionally, non-invasive alternatives like focused ultrasound (pFUS) or transdermal optogenetics—requiring minimal training—could bypass stimulation of implantable devices and reducing surgical expertise gaps[70].
Regenerative and remodeling neuroengineering
Long-term device efficacy is compromised by fibrotic encapsulation, electrode corrosion, and nerve damage. Biohybrid interfaces merging conductive biomaterials with regenerative therapies offer solutions. Electrodes coated with anti-inflammatory hydrogels (e.g., dexamethasone-eluting polymers) could mitigate fibrosis, while optogenetic tools enable cell-type-specific stimulation without metallic implants. Concurrently, neurotrophic support—via localized delivery of GDNF or NGF—may promote nerve regeneration around implants, as demonstrated in diabetic neuropathy models where GLP-1 agonists enhanced axonal integrity[71]. Future devices should leverage biodegradable electronics that dissolve after restoring endogenous glucose regulation, eliminating surgical extraction risks.
Design principles for future clinical trials
Earlier attempts at using peripheral neuromodulation for regulating glucose were restricted due to tiny sample quantities, brief periods, and diverse participant traits which held back the dependability and applicability of the results. To get past these issues, future randomized controlled experiments ought to utilize multicenter, double-blind, sham controlled setups with lengthened follow-up duration (at least 6 months) and sufficiently large sample amounts to spot both immediate and long-lasting metabolic impacts. These studies should include thorough main endpoints like decreasing HbA1c, TIR and CV together with secondary consequences such as HOMA-IR, GIR, inflammatory signs, autonomic function measures and outcomes reported by patients. Stratification in accordance with the type of diabetes, the length of the disease, the severity of neuropathy and the baseline body mass index would guarantee the identification of subgroups that respond, making it possible for precision medicine methods to be employed and through incorporating strict methodological controls, long-term monitoring and evaluations of biomarkers related to mechanisms. These randomized controlled trials would offer strong evidence regarding the effectiveness and safety of neuromodulation founded on the PNS, which in the end would promote its application in clinical practice.
CONCLUSION
PNS-mediated regulation of glucose homeostasis is an emerging and promising therapeutic paradigm. Evidence from preclinical and early clinical studies shows that autonomic and sensory pathways modulate hepatic glucose output, islet hormone secretion, and peripheral glucose uptake via organ-specific neuroendocrine circuits. Neuromodulation technologies such as VNS and focused ultrasound provide feasible, non-pharmacological intervention routes, yet translation to robust and sustained clinical benefit remains limited by stimulation specificity, neuropathy, and individual variability. Future strategies integrating mechanistic targeting, personalized protocols, and closed-loop control may overcome current barriers, enabling PNS-based interventions to become safe, effective, and scalable tools for managing diabetes and related metabolic disorders.
ACKNOWLEDGEMENTS
We thank the reviewers and editorial team for their insightful comments and the staff of Nanjing University of Chinese Medicine for technical support.
Footnotes
Provenance and peer review: Invited article; Externally peer reviewed.
Peer-review model: Single blind
Specialty type: Endocrinology and metabolism
Country of origin: China
Peer-review report’s classification
Scientific Quality: Grade B, Grade B, Grade B, Grade B
Novelty: Grade B, Grade B, Grade B
Creativity or Innovation: Grade B, Grade B, Grade B
Scientific Significance: Grade B, Grade B, Grade B
P-Reviewer: Chang KL, MD, PhD, China; Horowitz M, MD, PhD, Professor, Australia; Vyshka G, MD, PhD, Professor, Albania S-Editor: Lin C L-Editor: A P-Editor: Xu ZH
Lu JY, Ou HY, Wu CZ, Yang CY, Jiang JY, Lu CH, Jiang YD, Chang TJ, Chang YC, Hsieh ML, Wu WC, Li HY, Du YF, Lin CH, Hung HC, Tien KJ, Yeh NC, Lee SY, Yu HI, Chuang LM. A randomized trial on the effect of transcutaneous electrical nerve stimulator on glycemic control in patients with type 2 diabetes.Sci Rep. 2023;13:2662.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 1][Reference Citation Analysis (0)]
Lembo G, Vecchione C, Iaccarino G, Trimarco B. The crosstalk between insulin and the sympathetic nervous system: possible implications in the pathogenesis of essential hypertension.Blood Press Suppl. 1996;1:38-42.
[PubMed] [DOI]
Joly-Amado A, Soty M, Philippe E, Lacombe A, Castel J, Pillot B, Vily-Petit J, Zitoun C, Mithieux G, Magnan C. Portal Glucose Infusion, Afferent Nerve Fibers, and Glucose and Insulin Tolerance of Insulin-Resistant Rats.J Nutr. 2022;152:1862-1871.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 3][Reference Citation Analysis (0)]
Park CB, Lee CH, Cho KW, Shin S, Jang WH, Byeon J, Oh YR, Kim SJ, Park JW, Kang GM, Min SH, Kim S, Yu R, Kim MS. Extracellular Cleavage of Microglia-Derived Progranulin Promotes Diet-Induced Obesity.Diabetes. 2024;73:2009-2021.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 3][Reference Citation Analysis (0)]
Cotero V, Graf J, Miwa H, Hirschstein Z, Qanud K, Huerta TS, Tai N, Ding Y, Jimenez-Cowell K, Tomaio JN, Song W, Devarajan A, Tsaava T, Madhavan R, Wallace K, Loghin E, Morton C, Fan Y, Kao TJ, Akhtar K, Damaraju M, Barenboim L, Maietta T, Ashe J, Tracey KJ, Coleman TR, Di Carlo D, Shin D, Zanos S, Chavan SS, Herzog RI, Puleo C. Stimulation of the hepatoportal nerve plexus with focused ultrasound restores glucose homoeostasis in diabetic mice, rats and swine.Nat Biomed Eng. 2022;6:683-705.
[RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)][Cited by in Crossref: 50][Cited by in RCA: 48][Article Influence: 12.0][Reference Citation Analysis (0)]
Ashe J, Graf J, Madhavan R, Wallace K, Cotero V, Abate S, Pandey RK, Herzog R, Porindla SN, Shoudy D, Fan Y, Kao TJ, Puleo C. Investigation of liver-targeted peripheral focused ultrasound stimulation (pFUS) and its effect on glucose homeostasis and insulin resistance in type 2 diabetes mellitus: a proof of concept, phase 1 trial.QJM. 2023;116:667-685.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 8][Reference Citation Analysis (0)]
Kaniusas E, Kampusch S, Tittgemeyer M, Panetsos F, Gines RF, Papa M, Kiss A, Podesser B, Cassara AM, Tanghe E, Samoudi AM, Tarnaud T, Joseph W, Marozas V, Lukosevicius A, Ištuk N, Šarolić A, Lechner S, Klonowski W, Varoneckas G, Széles JC. Current Directions in the Auricular Vagus Nerve Stimulation I - A Physiological Perspective.Front Neurosci. 2019;13:854.
[RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)][Cited by in Crossref: 140][Cited by in RCA: 204][Article Influence: 29.1][Reference Citation Analysis (0)]