Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
June-2025 Volume 55 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
June-2025 Volume 55 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review)

  • Authors:
    • Ming-Wei Liu
    • Shao-Xin Duan
    • Xue-Yan Zhao
    • Qiong-Fen Wang
    • Shan-Lan Yang
    • Ni Ma
    • Xuan Li
  • View Affiliations / Copyright

    Affiliations: Department of Emergency Medicine, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Trauma, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Endocrinology, First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China, Department of Gastroenterology, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Oncology, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Pharmacy, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Rehabilitation, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
    Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 94
    |
    Published online on: April 17, 2025
       https://doi.org/10.3892/ijmm.2025.5535
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Sepsis‑induced organ dysfunction syndrome (ODS) arises from a dysregulated response to infection, leading to multiple life‑threatening organ dysfunctions, and is a common complication in critically ill patients. Sepsis results in varying degrees of injury to the brain, lungs, kidneys and liver, culminating in immune dysfunction and multiple ODS (MODS). Current evidence indicates a direct correlation between the severity of organ injury and the prognosis of septic patients. Understanding the mechanisms of MODS in sepsis and developing effective management strategies are vital research areas. The protective effects of dexmedetomidine (DEX) on sepsis are well established, demonstrating its capacity to mitigate injuries to the brain, lungs, kidneys, liver and immune system. The present study reviews recent research progress on the role and mechanisms of action of DEX in the treatment of sepsis.

Introduction

Sepsis-induced multiple organ dysfunction syndrome (MODS) involves dysfunction of two or more organs triggered by a dysregulated response to infection and is characterized by a series of physiological, pathological and biochemical abnormalities caused by infection (1) (Fig. 1). During the early stage of sepsis, inflammation is activated, leading to the release of numerous proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1) and IL-6, and inflammatory mediators, such as high-mobility group protein B1 and prostaglandins, resulting in immune overactivation. Additionally, immune cells, including T lymphocytes, release various cytokines, accompanied by redox imbalance. These mechanisms collectively activate endothelial cells, increasing the release of cytokines, nitric oxide (NO), platelet-activating factor and coagulation factors, thereby exacerbating sepsis and advancing to MODS, ultimately resulting in patient mortality (2) (Fig. 1). Despite extensive research on the pathogenesis and therapeutic targets of MODS, the incidence and mortality rates have improved little.

Figure 1

Mechanism of multiple organ dysfunction caused by sepsis. The endotoxins produced by pathogenic microorganisms stimulate inflammatory cells, which produce a large concentration of inflammatory mediators, leading to sepsis, septic shock and multiple organ dysfunction. Moreover, inflammatory mediators can lead to mitochondrial dysfunction, an oxidative stress response, coagulation dysfunction and microvascular blockage and exacerbate damage to multiple organs, such as the liver, brain and lungs. PGE2, prostaglandin E2; NO, nitric oxide; PAF, platelet-activating factor; ROS, reactive oxygen species.

Dexmedetomidine (DEX), a potent selective α2-adrenergic receptor agonist, functions as a sympathetic blockade within the nervous system and provides anxiolytic, sedative, analgesic and hemodynamic stabilizing effects (3). It induces sedation without causing respiratory depression, which is significant in clinical treatment. DEX is commonly used for preanesthetic induction and as a sedative in intensive care units (ICUs) (3,4). Previous studies have indicated that DEX has protective effects on various disease models: i) DEX enhances the 2-year survival of elderly patients in ICUs and improves cognitive function and quality of life in 3-year survivors (5); ii) DEX mitigates apoptosis, necrosis and autophagy in the brain and surrounding tissues in both in vitro and in vivo models (6); iii) DEX promotes cell survival by inhibiting apoptosis and protecting against bilirubin-related lung injury (7); iv) DEX protects against cerebral ischemia-reperfusion injury by inhibiting autophagy (7); and v) DEX shields against sepsis-related organ injury (8-11). The roles of DEX in mitigating lung, kidney, liver, brain and immune dysfunctions by regulating signaling pathways related to inflammation, apoptosis, pyroptosis, autophagy, and ferroptosis in sepsis treatment are summarized in Fig. 2.

Figure 2

Protective mechanism of dexmedetomidine against multiorgan dysfunction. LPS regulate tissue cell inflammation by modulating the MAPK, TLR4, or HMGB1/NF-κB signaling pathways; regulate tissue cell oxidative stress by modulating Nrf2/HO-1, P13K/Akt and NOX4 signaling pathways; and regulate cellular autophagy by modulating the P13K/Akt/mTOR signaling pathways. In addition, lipolysis can lead to endothelial damage, abnormal coagulation function and immune imbalance. Lipopolysaccharides induce multiple organ dysfunction through these mechanisms; however, dexmedetomidine alleviates lipopolysaccharide-induced multiple organ dysfunction by regulating these pathways. LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; TLR4, Toll-like receptor 4; HMGB1, high mobility group protein box-1; NLRP3, NOD-like receptor family pyrin domain containing 3; NF-κB, Nuclear factor κB; CAT, catalase; HO-1, heme oxygenase 1; ASC, apoptosis-associated speck-like protein; NCOA4, nuclear receptor coactivator 4; GSH, glutathione; GPX4, glutathione peroxidase 4; ROS, reactive oxygen species; SOD, superoxide dismutase; GSDMD, gasdermin D.

Epidemiology of sepsis and multiple organ dysfunction

Sepsis, a severe disease that threatens human health, is not only a medical issue but also a public health problem. A search of the World Health Organization website revealed that, according to data released in 2020 (12), there are 48.9 million cases and 11 million sepsis-related deaths globally, accounting for nearly 20% of all global deaths. Of these, nearly half of the sepsis cases occur in children, with an estimated 20 million child cases and 2.9 million deaths among children under the age of 5. The incidence and mortality rates of sepsis vary by region, with ~85% of sepsis cases and sepsis-related deaths occurring in low- and middle-income countries. Sepsis treatment is costly, and in high-income countries, the average hospital cost for sepsis exceeds $32,000 per patient (13). The incidence of sepsis in mainland China is similar to that in developed countries. However, owing to the large population base and relatively low ICU bed-to-population ratio, more patients with sepsis are not admitted to the ICU because of insufficient bed availability. A population-based epidemiological study of hospitalized sepsis patients conducted by Zhou et al (14) revealed that only 20% of patients with severe sepsis and 59% of patients with septic shock were admitted to the ICU. The burden of sepsis in mainland China may thus be underestimated. The study also revealed that the standardized incidence rate of sepsis is 461/10 million, with a mortality rate of 79/10 million. The high incidence and mortality of sepsis, as well as the annual medical burden it causes, highlight the need to improve the diagnosis and treatment of sepsis and, at the macrolevel, strengthen the management of sepsis prevention and treatment.

Sepsis-related organ injury or dysfunction is a common complication of sepsis, and in severe cases, it can lead to multiple organ dysfunction or failure, resulting in a poor prognosis (15). According to the American Society of Critical Care Medicine, the incidence of MODS in the United States is ~240-300/100,000 people, with ~54% of ICU-admitted patients developing MODS (16). The pathophysiology of MODS is complex, with an unclear pathogenesis and rapid disease progression, and the mortality rate is as high as ~30-40% (14). Studies have reported that the average mortality rate for individuals with two organ dysfunctions is 59%; for those with three organ dysfunctions, the average mortality rate is 75%; and for those with four or more organ dysfunctions, the average mortality rate is 100% (17). In terms of the frequency of organ dysfunctions occurring in MODS, the highest incidence is respiratory dysfunction, followed by gastrointestinal and renal dysfunction. Among them, renal dysfunction has the highest mortality rate, with an average of 79%, followed by respiratory dysfunction at 68%, gastrointestinal dysfunction at 59%, hepatic dysfunction at 55% and coagulation dysfunction at 44% (18). If severe infection is present, the mortality rate increases significantly. In recent years, the mortality rate of MODS has significantly increased with the number of organ failures and the age of patients, making MODS a major clinical challenge that urgently needs to be addressed in medicine.

Pharmacokinetics of DEX

The chemical molecular formula of DEX is C13H16N2, and its chemical name is (+)-4-(2,3-dimethylphenyl)-ethyl-1H-imidazole (19). DEX shares a similar chemical structure with clonidine, including an imidazole ring and a benzene ring, indicating that both can act as agonists on α-adrenergic receptors. As a highly selective α2-adrenergic receptor agonist, DEX has a binding affinity ratio of 1,620:1 for α2-adrenergic receptors compared with α1 receptors. Its affinity for α2-adrenergic receptors is 8-fold greater than that of clonidine. Studies have shown that the pharmacokinetics of DEX are strongly predictable (20). For example, during absorption, owing to the first-pass effect, the oral bioavailability of DEX is extremely low. The bioavailability is only 16% for sublingual administration, whereas intranasal administration achieves a bioavailability of 84%. For intramuscular and subcutaneous injections, the bioavailabilities are 73 and 51%, respectively. Therefore, intranasal, intravenous and intramuscular routes of administration are all applicable. However, considering the storage and usage of DEX, along with its physicochemical property of being fully soluble in water at a pH of 7.4, clinical formulations are commonly provided as colorless, clear liquid hydrochloride injection solutions. Intranasal administration is particularly beneficial for special populations, such as children and elderly individuals (21). DEX is a highly plasma protein-bound drug, with a binding rate of 94% to plasma albumin and α1-glycoprotein. Studies have shown that the plasma protein binding rate of DEX is not significantly influenced by sex or age (22). Additionally, DEX has a shorter half-life than does clonidine. Its distribution half-life is ~6 min, allowing it to be distributed rapidly in the body following intravenous injection. The elimination half-life (t1/2) is ~2 h, with intravenous infusion lasting up to 24 h. The linear pharmacokinetic range of DEX in the body is 0.2-0.7 µg/kg·h. It is metabolized primarily in the liver, with the metabolites being excreted mainly through urine (95%) and feces (5%) (21).

Pharmacodynamics of DEX

DEX is a potent and highly selective α2-adrenergic receptor agonist that exerts various effects by stimulating three subtypes of α2 receptors in the body (α2A, α2B and α2C). For example, the activation of α2A receptors, which are located primarily in the locus coeruleus of the brain, can produce effects such as sedation, analgesia, hypnosis and neuroprotection; the activation of α2B receptors, which are distributed mainly in the thalamus, heart, liver, spleen and aorta, can lead to analgesia, peripheral arterial vasoconstriction, and the suppression of centrally mediated shivering; and the activation of α2C receptors, which are found in the heart, spleen and aorta, can result in cognitive and emotional changes (3). Therefore, by stimulating α2 receptors in these organs and tissues, DEX can exert multiple biological effects, including sedation, analgesia, hypnosis, maintenance of normal respiration, and a reduction in organ damage.

DEX induces a unique sedative response characterized by a smooth transition from sleep to wakefulness, allowing patients to communicate and interact when stimulated by external factors. It mimics the effects of 'natural sleep', enabling patients to remain in a sleep-like state during surgery while being easily arousable. DEX has minimal toxic side effects on the body and, regardless of the dosage, does not cause respiratory depression (23). Sleigh et al (24) suggested that the sedative and hypnotic effects of DEX are mediated by the activation of pre- and postsynaptic α2 receptors in the central nucleus of the brainstem locus coeruleus, which function through endogenous physiological sleep pathways to exert sedative and hypnotic effects. Clinical studies have shown that DEX has dose-dependent sedative effects, with doses of 0.2-0.6 µg/kg·h inducing significant yet easily reversible sedation. At sufficiently high doses, DEX can produce deep sedation and even general anesthesia, indicating its potential role as part of total intravenous anesthesia. Although the U.S. Food and Drug Administration has approved DEX for short-term sedation (<24 h), multiple clinical studies have confirmed that continuous use of DEX for several days can mitigate adverse effects in patients with ICU (3,23). Ding et al (25) reported that, compared with traditional sedatives, long-term ICU sedation with DEX reduced mechanical ventilation time of patients by 22%, shortened hospital stays by 14%, and did not significantly affect mortality. When the sedative effects of midazolam, propofol and DEX in mechanically ventilated ICU sepsis patients were compared, DEX demonstrated comparable sedation efficacy while better stabilizing blood pressure and cardiac function. Kawazoe et al (26), in a study of 201 mechanically-ventilated patients with sepsis, reported that the sedation control rate during mechanical ventilation was significantly greater in the DEX group than in the non-DEX group. Furthermore, DEX significantly reduced the levels of inflammatory markers such as C-reactive protein and procalcitonin, alleviating systemic inflammatory responses.

Pharmacokinetic study of DEX in special populations

Inside the child's body

DEX can be administered via intramuscular injection, intravenous injection, nasal drops, or oral administration inside the child's body. The use of nasal drops or oral administration can prevent high peak plasma levels that may cause respiratory and circulatory depression after intravenous injection. It also helps avoid the irritation caused by intravenous administration, rendering it more suitable for children. The nasal mucosa has a rich distribution of aqueous channels, providing high drug permeability and rapid absorption. It also bypasses the hepatic first-pass effect. Research by Miller et al (27) indicates that nasal administration of DEX for pediatric sedation can provide adequate blood drug concentrations, with a bioavailability >80%, whereas oral administration has a bioavailability of only 16% owing to the first-pass effect (20). Yadav and Ramdas (28) conducted a randomized controlled trial of intranasal DEX for preoperative pediatric anesthesia. The results showed that, compared with midazolam, it offered more stable hemodynamics, a shorter time to achieve sedation, and lower anxiety scores. It is an excellent sedative and anxiolytic and can be safely used in children. DEX is a drug with a strong binding ability to proteins, with a protein binding rate of 94%. It is distributed quickly and extensively in the human body and is also a highly lipophilic drug that easily crosses the blood-brain barrier (29). Non-compartmental analysis has shown that the distribution half-life in healthy volunteers is ~6 min (30). Studies have indicated that in critically ill patients with hypoalbuminemia, long-term administration of DEX significantly increases its apparent Vd (31). In pediatric pharmacokinetics for children <11 years old, the results revealed that the steady-state Vd (Vss) is negatively correlated with the age of the subjects (32), indicating that younger children require a larger initial dose of DEX to reach a certain blood concentration. DEX is metabolized primarily in the liver, with most of its metabolites excreted through the kidneys and a small portion through feces (33,34). Research on the pharmacokinetics of DEX in neonates and infants shows that the drug clearance (CL) rate increases with age, particularly within the first 2 weeks after birth, whereas after 1 month of age, the impact of age on the CL rate is minimal (33). The metabolic enzyme system of newborns and infants is not fully mature, resulting in low CL rates. Research has shown that the expression and activity of cytochrome P450 enzymes approach adult levels by the age of 1 (35). The CL rate of full-term newborns is ~42.2% of that of adults, reaching 84.5% at 1 year of age (29). Premature infants have even lower CL rates [0.3 l/(h x kg) than 0.9 l/(h x kg)] and longer elimination half-lives (7.6 compared with 3.2 h) (33). The CL rate of DEX is only mildly affected by hypoproteinemia, but since the liver is the primary metabolic organ, the CL rate decreases as the severity of liver damage increases (32).

Among the elderly group

With increasing age, the body fat content also increases in the elderly group (36). For males, from the ages of 20 to 30, the fat content increases from 8% to 20%, whereas for females, it increases from 30 to 50% (37). First, lipophilic drugs (such as DEX) have a relatively large volume of distribution (Vd) (38). Second, the plasma protein concentration decreases with age, which in turn increases the free fraction of drugs with a high protein binding rate (39), thus increasing the Vd of DEX. Changes in liver and kidney function in elderly individuals can affect plasma protein binding rates. Additionally, an increase in fat content affects organ blood flow and the activity of mixed-function oxidases (40). Studies on the population pharmacokinetics of ICU patients receiving long-term DEX infusion have revealed that age and plasma protein concentration can influence the pharmacokinetic characteristics of DEX. In the elderly population, CL decreases, and the t1/2 increases. Moreover, patients with low plasma ALB concentrations have an increased Vd and a prolonged t1/2. The CL of the elderly population, with an average age of 80 years, is 25% lower than that of those aged 60 years (41). This finding is consistent with the report by Venn et al (42). CL is related primarily to liver blood flow and is less affected by the protein binding rate (43). As various bodily functions decline in elderly individuals, the CL of DEX decreases, leading to an extended t1/2. Lin et al (44) conducted a population pharmacokinetic study on post-operative ICU patients, and the results revealed that factors such as age and sex had no impact on their pharmacokinetics. This may be due to the small sample size and the high variability in the pharmacokinetic parameters of DEX (45). Moreover, the aforementioned study did not provide standard errors or confidence intervals, indicating insufficient model stability and reliability.

In individuals with liver dysfunction

Individuals with liver dysfunction, the CL of DEX decreases, with significant individual variability (46). According to the Child-Pugh classification, liver dysfunction patients are divided into three groups: mild (n=5), moderate (n=6), and severe (n=5). Compared with those in the three control groups, the average CL rates of DEX were 41, 49 and 68% lower in patients with mild, moderate and severe liver dysfunction, respectively. Compared with those in both the mild group and the control group, the Vss and t1/2 were significantly greater in severe liver dysfunction patients (P<0.05) (47), indicating that when DEX is used in patients with liver dysfunction, dose adjustments should be considered.

In individuals with renal insufficiency

There was no statistically significant difference in the pharmacokinetic parameters of DEX in patients with renal insufficiency compared with healthy subjects. A comparative study was conducted between 6 patients with renal insufficiency (creatinine CL <30 ml/min) and 6 healthy volunteers, and the results revealed that there were no statistically significant differences in maximal plasma/serum concentration, time to maximal plasma/serum concentration, area-under-the-curve plasma/serum concentrations, CL, or Vss (48). However, in patients with renal insufficiency, the elimination-phase concentration was lower, and elimination was faster, with a 17% decrease at t1/2 (P<0.05). In patients with renal insufficiency, the protein binding rate of alfentanil, which has a high plasma protein binding rate, decreases from 19 to 11% (49). Therefore, the decrease in the t1/2 of DEX may also be due to the reduced plasma protein binding rate in patients with renal insufficiency, leading to an increase in the amount of drug entering the elimination phase.

Others

There have been few studies on the pharmacokinetics of DEX in other special populations (including pregnant women, parturient women and lactating patients). In one case, a pregnant woman with spinal muscular atrophy who underwent routine cesarean section had DEX infusion stopped, and 68 min later, the fetus was delivered. DEX was detected in the umbilical blood, suggesting that DEX can cross the placental barrier (50). DEX can cause a significant increase in blood glucose levels in both the mother and the fetus (51).

Organ protection mechanisms of DEX

In recent years, an increasing number of basic and clinical studies have shown that DEX may exert organ-protective effects through anti-inflammatory, antiapoptotic, antioxidative stress, and autophagy-regulating mechanisms involving the heart, brain, lungs, kidneys, liver and gastrointestinal tract. Despite its common effects, the biological mechanisms of DEX vary across different organs. Studies have shown that perioperative infusion of DEX can reduce the levels of inflammatory factors such as interleukin-6 (IL-6), IL-8, and TNF-α, with the effect being more significant during continuous intraoperative infusion than during single-dose administration (52). The anti-inflammatory mechanism of DEX is closely related to the inhibition of the nuclear factor κB (NF-κB) pathway and Toll-like receptors via α2 receptors (53). The vagus nerve and nicotinic acetylcholine receptors are key components of the cholinergic anti-inflammatory pathway. In addition to acting directly on α2 receptors, DEX can activate the cholinergic anti-inflammatory pathway to exert its anti-inflammatory effects (54,55). DEX also has antiapoptotic effects on various cell types, potentially through activation of the α2AR/PI3K/Akt and p38MAPK/ERK signaling pathways (56-58). Additionally, DEX may inhibit mitochondrial apoptosis by activating the JAK2/STAT3 signaling pathway via the long non-coding RNA HCP5 (59). However, some studies have shown that high concentrations of DEX can induce hippocampal neuronal apoptosis. In vitro experiments suggest that DEX concentrations ≥100 µmol/l significantly reduce cell viability and induce neuronal apoptosis (60). In different cell injury models, such as vascular smooth muscle cells, renal tubular epithelial cells and hepatocytes, DEX can alleviate oxidative stress by inhibiting the production of malondialdehyde (MDA) and reactive oxygen species (ROS) and increasing the activity of superoxide dismutase (SOD) (61-63). DEX also inhibits the decrease in the mitochondrial membrane potential and reduces respiratory chain enzyme complex damage, thereby suppressing the mitochondrial oxidative stress induced by general anesthesia (64). Autophagy is an adaptive catabolic process that maintains intracellular homeostasis by engulfing intracellular substances, including damaged organelles, unfolded proteins and pathogens. In cases of damage to critical organs such as the heart, lungs, brain and kidneys, DEX treatment can increase autophagy levels (65,66). However, studies have also shown that DEX can suppress excessive autophagy in cardiomyocytes and neurons to protect organs (67,68). Therefore, under different pathological conditions in various organs, DEX has distinct regulatory effects on autophagy.

Mechanism of MODS induced by sepsis

Currently, MODS caused by sepsis is considered to be associated with factors such as uncontrollable infection, the systemic inflammatory response, immune paralysis, coagulation activation, microcirculatory failure and hypoxia (69). Studies have shown that immune-metabolic reprogramming occurs at various stages of sepsis, making it a key regulatory factor in the pathogenesis of this disease (70). Polymorphonuclear neutrophils (PMNs) are innate immune cells that respond first to infectious pathogens and play a crucial role in host defense. During infection, the body reacts rapidly to local inflammatory stimuli, recruiting numerous PMNs efficiently from the bloodstream to the site of inflammation. PMNs eliminate pathogens through phagocytosis, degranulation, respiratory bursts, the generation and release of neutrophil extracellular traps (NETs), and the production of inflammatory factors such as TNF-α, IL-1β, IL-6 and IL-8, thus controlling infection (71). Under normal conditions, inflammatory responses are tightly regulated and confined to the infection site. After infection is resolved, inflammation subsides, and PMNs undergo apoptosis and are cleared. However, during sepsis, systemic inflammation causes PMN polarization. The chemotaxis and migration functions of PMNs are impaired, leading to a reduced number of PMNs at the infection site (source site) and increased PMN infiltration in distant vital organs (72). Activated PMNs in organs release proinflammatory cytokines, ROS, lysosomal enzymes, NETs and other active substances, resulting in cell, tissue and organ damage and ultimately leading to MODS (73). A previous study has reported that ROS produced by polarized PMNs during sepsis can damage various organs, including the heart, kidneys, liver and lungs, through multiple mechanisms, contributing to the development of MODS (74). Moreover, activated PMNs are significant sources of matrix metalloproteinases (MMPs), particularly MMP-9, which can degrade extracellular matrix proteins such as elastin and collagen. This degradation can stimulate the immune system and create a positive feedback loop of tissue degradation and immune cell infiltration, further exacerbating organ damage at infiltration sites (75). Substances released by PMNs also stimulate the production of NO by various cells, leading to blood pressure reduction and the generation of peroxynitrite, which causes cellular damage. In sepsis-induced myocardial injury, peroxynitrite can alter protein structure and function, causing PMNs to adhere to and accumulate in the endothelium and myocardium, ultimately resulting in cardiovascular dysfunction (76). Additionally, ROS, NETs and lysosomal enzymes released by polarized PMNs can act as damage-associated molecular patterns (DAMPs), causing endothelial injury and increased vascular permeability. DAMPs can also activate immune cells, including macrophages, triggering uncontrolled inflammatory responses. The release of inflammatory factors results in the recruitment of more PMNs, further stimulating the excessive production of NETs and ROS and creating a positive feedback loop that exacerbates organ damage. These harmful substances contribute to vascular injury and death in organs, eventually leading to organ failure. During the development of sepsis-induced lung injury, increased PMN infiltration promotes bacterial translocation in the lungs, exacerbating ROS production and worsening lung damage (77). The progression of MODS in sepsis significantly increases patient mortality, making the protection of organ function a critical strategy for improving sepsis outcomes, with the regulation of immune cell function being a key factor.

Protective mechanism of DEX on important organs

Current research indicates that DEX has a protective effect on sepsis-induced multiorgan dysfunction, as follows:

Lung injury

Sepsis often leads to multiple organ injuries, with lung injury being particularly prevalent, occurring in 83-100% of cases (78). The lipopolysaccharide (LPS)-induced inflammatory response and oxidative stress through the regulation of inflammatory pathways such as MAPK and TLR4, as well as the oxidative stress pathway Nrf2/heme oxygenase-1 (HO-1), are the primary causes of lung injury, and sepsis-induced alveolar cell pyroptosis and ferroptosis through the regulation of pyroptosis pathways such as the NLRP3 pathway, as well as the ferroptosis pathway SLC7A11/glutathione peroxidase 4 (GPX4), are also involved in acute lung injury (Fig. 3) (78). Research has shown that inhibiting the high mobility group protein box-1 (HMGB1)/myeloid differentiation factor-88 (MyD88)/NF-κB pathway and NLRP inflammasome activation induced by cecal ligation and puncture (CLP) can reduce CLP-induced lung inflammation and alveolar cell pyroptosis and ameliorate sepsis-induced acute lung injury (79), and increasing the activity of the Nrf2/HO-1 pathway can reduce LPS-induced oxidative stress and inflammation in alveolar cells and ameliorate sepsis-induced acute lung injury (80,81). Therefore, inhibitors of sepsis-induced alveolar cell inflammation, oxidative stress, pyroptosis and ferroptosis may help improve sepsis-induced lung injury. Wu et al (82) demonstrated that DEX inhibits the expression of Toll-like receptor 4 (TLR4), a molecule associated with endotoxin recognition, and the activation of the nuclear transcription factor NF-κB. This inhibition reduces TNF-α and IL-6 levels in alveolar lavage fluid and plasma, significantly decreasing the fatality rate in CLP-induced septic rats within 24 h. Additionally, DEX attenuates LPS-induced acute lung injury in rats by inhibiting the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/forkhead box O1 (FoxO1) signaling pathway and mitigating oxidative stress, mitochondrial dysfunction and apoptosis (83). DEX also reduces oxidative stress and inflammation in alveolar cells and maintains the balance of mitochondrial dynamics through the HIF-1α/HO-1 signaling pathway, thereby ameliorating endotoxin-induced acute lung injury both in vivo and in vitro (84). Furthermore, DEX inhibits the secretion of inflammatory mediators such as TNF-α, IL-6, IL-1β and NO by alveolar cells via the HMGB1-mediated TLR4/NF-κB and PI3K/Akt/mammalian target of rapamycin (mTOR) pathways, increases the production of the antioxidant stress markers SOD and glutathione peroxidase, and alleviates LPS-induced lung injury by reducing inflammation and oxidative stress (85). A previous study indicated that DEX maintains endothelial barrier integrity by reducing angiopoietin 2 (ANG2) levels and increasing endothelial VE-cadherin in rats with CLP-induced sepsis, thereby mitigating lung inflammation (86,87). The Ang-tyrosine kinase receptor 2 (Tie2) axis is a well-studied pathway that regulates endothelial barrier function. Ang1 binds to and activates angiopoietin Tie2 receptors, upregulating VE-cadherin to stabilize the cell barrier. Conversely, ANG2 competes with Tie2 receptors, antagonizing the anti-inflammatory effects of Ang1, leading to barrier damage and endothelial cell activation, which induces inflammation (87). DEX reduces the wet/dry weight ratio of lung tissue, leukocyte infiltration, plasma ANG2 levels and ANG2/ANG1 ratios and decreases VE-cadherin phosphorylation through the ANG1-Tie2-VE-cadherin signaling pathway, thereby alleviating endothelial barrier damage and lung injury in septic rats (87). Additionally, DEX ameliorates sepsis-induced acute lung injury by modulating the adenosine monophosphate-activated protein kinase (AMPK)/silent information regulator 1 (SIRT1) pathway (88). These data suggest that DEX improves sepsis-induced lung injury by inhibiting multiple proinflammatory and oxidative stress pathways. However, these studies remain in the animal experimentation stage, and further research is needed to explore their clinical applications.

Figure 3

Mechanisms of sepsis-induced lung injury. LPS regulates oxidative stress and mitochondrial dysfunction by modulating the Nrf2/HO-1 signaling pathways; regulates lung inflammation by modulating the MAPK, TLR4 or HMGB1/NF-κB signaling pathways; regulates lung inflammation by modulating the MAPK or TLR4 or HMGB1/NF-κB signaling pathways; and regulates alveolar cell pyroptosis by modulating the NLRP3/caspase-1 signaling pathway and regulates alveolar cell ferroptosis by modulating the NF-κB/ROS/GPX4 signaling pathway. LPS, lipopolysaccharide; Nrf2, nuclear factor erythroid 2-related factor 2; HO-1, heme oxygenase 1; ROS, reactive oxygen species; MAPK, mitogen-activated protein kinase; TLR4, Toll-like receptor 4; HMGB1, high mobility group protein box-1; NLRP3, NOD-like receptor family pyrin domain containing 3; NF-κB, nuclear factor κB.

Kidney injury

The kidney is a primary target organ in sepsis (89). Acute kidney injury (AKI) manifests as progressively worsening renal function, severe oliguria and fluid overload, often necessitating renal replacement therapy. This condition not only increases hospitalization and treatment costs but also severely impacts patient prognosis. The pathophysiological process of sepsis-induced AKI remains poorly understood, and effective prevention and treatment measures are lacking in clinical settings. Studies have revealed that the pathogenic mechanisms of sepsis-induced AKI are complex, involving LPS-induced inflammation, microcirculatory disorders and oxidative stress by regulating inflammatory pathways, such as TLR4 and MAPK, as well as oxidative stress-related pathways, such as Nrf2, NADPH oxidase 4 (NOX4) and P13k/Akt, with inflammation being a pivotal factor (Fig. 4) (89,90). Research has shown that inhibiting the activity of the TLR4/MyD88/NF-κB signaling pathway can inhibit LPS-induced kidney inflammation and improve sepsis-induced AKI in mice (91) and that decreasing NLRP3 inflammasome activation can alleviate LPS-induced tubular cell inflammation and pyroptosis and ameliorate LPS-induced kidney injury (92). The inhibition of NOX4 protects against sepsis-induced AKI by reducing the generation of ROS and the activation of NF-κB signaling, which suppresses mitochondrial dysfunction, oxidative stress, inflammation and apoptosis (93). Therefore, inhibiting inflammation and oxidative stress and improving renal microcirculation may help alleviate sepsis-induced AKI. Liu et al (94) investigated the immunomodulatory effects of DEX on sepsis-related AKI induced by tail vein injection of LPS in rats at doses of 3, 5, 10 and 20 µg/kg iv. DEX was found to reduce inflammatory cytokine levels in a dose-dependent manner. The addition of the α2 adrenergic receptor antagonist yohimbine negated the regulatory effect of DEX on cytokine production, confirming its anti-inflammatory role as an α2 adrenergic receptor agonist. Jin et al (95) established a sepsis-related AKI model in rats via LPS induction. The DEX group received 80 mg/kg DEX daily starting 4 days before modeling. The results indicated that DEX downregulates TLR4 protein expression, inhibits MyD88 and NF-κB activation, and causes expression of inducible NO synthase (iNOS), thereby suppressing inflammatory factor production. These findings indicate that DEX may reduce inflammation by inhibiting the TLR4/MyD88/NF-κB/iNOS signaling pathway and attenuating LPS-induced NLRP3 inflammasome activation through the TLR4/NOX4/NF-κB pathway, thereby mitigating oxidative stress (96). Wang et al (97) investigated an inflammatory proximal tubular epithelial cell model and an LPS-induced sepsis-related AKI model in mice. The results revealed that DEX significantly increased SOD activity in renal tissue, decreased the production of the lipid peroxidation product MDA, inhibited ROS production in renal tubular epithelial cells, and ameliorated sepsis-induced AKI. Zhao et al (98) pretreated an LPS-induced sepsis-related AKI model in rats with ip DEX at 30 µg/kg and discovered that DEX enhanced autophagy through α2 adrenergic receptors and inhibited the PI3K/Akt/mTOR pathway, leading to the CL of damaged mitochondria and a reduction in oxidative stress and apoptosis in LPS-induced AKI. The use of autophagy inhibitors, such as AT2 receptor-interacting proteins and 3-methyladenine, blocked the reno-protective effect of DEX, suggesting that regulating autophagy is crucial in the pathogenesis of LPS-induced AKI. By modulating renal autophagy, DEX improved oxidative stress and cell apoptosis. These studies demonstrate the protective effects of DEX on the kidney through various signaling pathways. However, since most experiments are based on animal models and the mechanisms of action have not yet been fully elucidated, further research is necessary.

Figure 4

Mechanisms of sepsis-induced kidney injury. LPS regulate kidney inflammation by modulating the NLRP3/NF-κB or TLR4/Myd88/NF-κB signaling pathways; regulate oxidative stress and ferroptosis by modulating the NOX4, Nrf2 or P13K/Akt signaling pathways; and LPS-induced inflammatory mediators can lead to renal microcirculation disorders, renal hypoperfusion, ischemia and hypoxia and exacerbate sepsis-induced kidney injury. LPS, lipopolysaccharide; PAMPS, pathogen-associated molecular patterns; DAMPs, damage-associated molecular patterns; NLRP3, NOD-like receptor family pyrin domain containing 3; TLR4, Toll-like receptor 4; MyD88, myeloid differentiation factor 88; PI3K, phosphatidylinositol 3-kinase; Akt, protein kinase B; NOX4, NADPH oxidase 4; ROS, reactive oxygen species; SOD, superoxide dismutase; GSH, glutathione; GPX4, glutathione peroxidase 4.

Liver injury

Endotoxin-induced liver failure is the second leading cause of multiple organ failure (99). Sepsis-induced hepatic injury resulting from endotoxin induces inflammation, hepatic blood flow alterations, microcirculatory disturbances and mitochondrial dysfunction by regulating inflammatory pathways, such as the MAPK and TLR4 pathways, and oxidative stress- and ferroptosis-related pathways, such as the Nrf2/HO-1 pathways (Fig. 5) (99,100). Hepatic sinusoidal cells are the initial sites of liver injury. The macrophages (KCs) within these sinusoids are pivotal in the inflammatory process and endotoxin-mediated hepatic injury (101). High doses of LPS provoked KC overactivation and reduced phagocytic capacity while increasing CD14 expression on KC surfaces. The LPS receptor/signaling complex, which is composed of TLR4, CD14 and MD2, further activates KCs, releasing a cascade of proinflammatory mediators that exacerbate endotoxin-induced hepatic damage (102). Thus, inflammation is central to sepsis-induced liver injury. Suppressing this inflammation may mitigate acute hepatic injury in sepsis. Studies on septic murine liver tissue revealed that DEX mitigates portal venous and sinusoidal congestion, reduces hepatic sinusoidal swelling, and decreases lymphocyte and neutrophil infiltration in the liver hilum, effectively curbing hepatic inflammation (103). DEX has been demonstrated to prevent acute hepatic injury in LPS-induced septic rats by inhibiting the TLR4/MyD88/NF-KB pathway. Additionally, DEX modulates caveolin-1, inhibits the TLR4/NLRP3 pathway, and curtails proinflammatory cytokine release, thus alleviating sepsis-induced hepatic injury (104,105). Furthermore, autophagy in the liver is crucial for maintaining cellular energy and nutrient balance, clearing damaged hepatic proteins, and counteracting oxidative stress (106). Research conducted by Yu et al (106) indicated that DEX potentially enhances autophagy via the SIRT1/AMPK pathway, mitigating inflammation in CLP-induced liver injury and increasing the survival rate of model mice by 20% within 24 h. LPS induces liver damage through apoptosis mediated by ROS. DEX activates the GSK-3β/MKP-1/Nrf2 pathway via the α2-adrenergic receptor, diminishing LPS-induced oxidative stress and apoptosis in the rat liver and thereby ameliorating sepsis-induced liver injury (107). DEX pretreatment facilitates M2 macrophage activation through PPARγ/STAT3 signaling, reducing liver inflammation and alleviating ischemia-reperfusion injury in mice (108). This evidence indicates that DEX confers protection against sepsis-induced liver injury by modulating inflammation, apoptosis, autophagy, and ferroptosis-related pathways and proteins. However, these findings are confined to animal research, and clinical validation is needed.

Figure 5

Mechanism of sepsis-induced liver injury. LPS regulate liver cell inflammation by modulating the MAPK/NF-κB and TLR4/NF-κB signaling pathways; regulate liver cell oxidative stress and ferroptosis by modulating the Nrf2/HO-1 signaling pathways; and LPS-induced inflammatory mediators can lead to endothelial cell damage, microcirculation disorders, adaptive pathway activity in liver cells, and exacerbated sepsis-induced liver injury. LPS, lipopolysaccharide; TLR4, Toll-like receptor 4; MyD88, myeloid differentiation factor 88; NLRP3, NOD-like receptor family pyrin domain containing 3; PI3K, phosphatidylinositol 3-kinase; Akt, protein kinase B; NOX4, NADPH oxidase 4; PPARγ, peroxisome proliferator-activated receptor gamma; ROS, reactive oxygen species; SOD, superoxide dismutase; GSH, glutathione; GPX4, glutathione peroxidase 4; NF-κB, nuclear factor-kappa B.

Myocardial injury

Epidemiological studies indicate that the global mortality rate of sepsis remains high and is gradually increasing (109). The heart is the organ most frequently affected during sepsis. It has been revealed that 40-60% of septic patients suffer from myocardial injury, with a fatality rate of 70-90%, which correlates positively with the severity of the injury (110). The extensive inflammation and 'cytokine storm' associated with sepsis lead to ischemia, oxidative stress, pyroptosis, ferroptosis, and hypoxic damage to myocardial cells by regulating pathways related to pyroptosis and inflammation, such as NLRP3 and MAPK, as well as pathways related to oxidative stress and iron death, such as Nrf2/HO-1 and NCOA4/GPX4 (Fig. 6), resulting in a significantly higher mortality rate than that in septic patients without myocardial injury. Long-term prognosis studies have also shown that survivors of sepsis-induced myocardial injury experience markedly reduced quality of life (111). Previous studies have identified that the activation of Nrf2/HO-1 and the inhibition of the NLRP3 signaling pathway can inhibit LPS-induced inflammation and oxidative stress; protect cardiomyocytes against LPS-induced injury (112); suppress NLRP3-mediated pyroptosis via the upregulation of the Nrf2/HO-1 signaling pathway in vivo and in vitro; decrease oxidative stress, inflammatory responses and NLRP3-mediated pyroptosis; and reduce LPS-induced myocardial injury (113,114). Thus, mitigating sepsis-related inflammation may alleviate myocardial injury and reduce mortality (115). In experimental mice treated with DEX, the incidence of septic shock was lower, and the plasma levels of the inflammatory factors TNF-a, IL-1β and IL-6 were significantly lower. Additionally, NF-кB binding activity was downregulated, increasing survival rates (116,117). In LPS-induced septic heart injury in rats, DEX appears to confer cardioprotective effects by diminishing the expression of NF-κB p65 and phosphorylated STAT3, thereby inhibiting the expression of the apoptotic proteins Caspase-3, Caspase-8, Bax and p53 and the inflammatory factors IL-6, IL-1β and TNF-α (118). DEX also inhibits GPX4 expression, upregulates HO-1, and reduces sepsis-induced cardiomyocyte ferroptosis and myocardial injury in mice (119). Compared with pre-surgery, the administration of varying doses of DEX to septic patients significantly decreased the levels of inflammatory factors such as IL-1, IL-6 and TNF-α within 24 h post-surgery. Moreover, high, medium, and low doses of DEX maintain and enhance cardiac function, significantly mitigating the adverse effects of inflammatory mediators on the heart (120). Despite substantial progress in understanding the myocardial protective effects of DEX, the underlying mechanisms remain incompletely elucidated, necessitating further research.

Figure 6

Mechanisms of sepsis-induced myocardial injury. LPS regulate cardiomyocyte inflammation by modulating the MAPK/NF-κB, MGB1/TLR4/NF-κB, and P13K/Akt/NF-κB signaling pathways; regulate liver cell oxidative stress and ferroptosis by modulating the Nrf2/HO-1 signaling pathways; regulate cardiomyocyte pyroptosis by modulating the NLRP3/caspase-1 signaling pathways; and regulate cardiomyocyte oxidative stress by modulating the Nrf2/HO-1, P13K/Akt, and NOX4 signaling pathways. In addition, LPS-induced inflammatory mediators can lead to calcium influx, and calcium overload in myocardial cells exacerbates sepsis-induced cardiac injury. LPS, lipopolysaccharide; TLR4, Toll-like receptor 4; MyD88, Myeloid differentiation factor 88; NLRP3, NOD-like receptor family pyrin domain containing 3; PI3K, phosphatidylinositol 3-kinase; Akt, protein kinase B; NOX4, NADPH oxidase 4; HO-1, heme oxygenase 1; ASC, apoptosis-associated speck-like protein; NCOA4, nuclear receptor coactivator 4; GSH, glutathione; GPX4, glutathione peroxidase 4; MAPK, mitogen-activated protein kinase; ROS, reactive oxygen species.

Gastrointestinal injury

Late-stage sepsis often progresses to MODS, a primary cause of mortality in ICU patients (121). As the body's largest 'bacteria reservoir' and 'endotoxin reservoir', the gastrointestinal tract is among the first and most severely impacted organs in the pathophysiological progression of sepsis (122). Sepsis results in the generation of substantial amounts of endotoxins and inflammatory mediators that compromise the gastrointestinal mucosal barrier, increase mucosal permeability, and promote intestinal bacterial translocation. Consequently, numerous bacteria and endotoxins infiltrate the bloodstream via lymphatic and capillary pathways, instigating a 'secondary infection' and triggering entheogenic endotoxemia. This can elicit unrestrained inflammatory responses, exacerbate intestinal damage, create a vicious cycle, and potentially advance to MODS, resulting in fatality (122,123). Hence, prompt evaluation of gastrointestinal function and early intervention in sepsis patients are essential to mitigate damage and improve patient prognosis. As shown in Table I (124-131), DEX ameliorates ischemia/reperfusion, LPS and CLP-induced intestinal injury in rats by suppressing TLR4, p38 MAPK/NF-κB SIRT3-dependent PINK1/HDAC3/p53, and alpha2AR/caveolin-1/p38MAPK/NF-κB signaling pathways in intestinal tissue; increasing ZO-1 and occludin expression; decreasing inflammatory cytokine levels and MLCK protein expression; and increasing intestinal permeability resulting from burns with concurrent histological damage. However, deep sedation with DEX, unlike propofol, significantly suppresses gastrointestinal motility in endotoxemic mice, an effect that dissipates after 24 h of sedation (131). These results indicate that while DEX has protective effects against intestinal barrier dysfunction, its administration in sepsis and intestinal paralysis requires careful consideration.

Table I

Study on the protective mechanism of DEX against intestinal injury caused by sepsis.

Table I

Study on the protective mechanism of DEX against intestinal injury caused by sepsis.

First author, yearObject of studyDosage of DEXEffectRegulatory mechanism of the intestine injury(Refs.)
Chen et al, 2015CLP-ICLP-induced intestinal injury5 µg × kg−1 × h−1Reducing systemic inflammation and alleviating CLP-induced intestinal tissue damageSuppressing TLR4 expression in intestinal tissue, curtailing inflammatory mediators(124)
Qin et al, 2021Burn-induced intestinal barrier damage5 µg × kg−1 × h−1Decrease in intestinal permeability and histological damage to the intestineAnti-inflammatory effect and protected tight junction complexes by inhibiting the MLCK/ p-MLC signaling pathways(125)
Yang et al, 2021Intestinal ischemia/ reperfusion injury5 µg × kg−1 × h−1Reduce intestinal I/R injury in rats and OGD/R damage in Caco-2 cellsAttributed to anti-inflammatory effects and inhibition of the TLR4/MyD88/NF-κB signaling pathway(126)
Ye et al, 2022DSS-induced colitis mice5 µg × kg−1 × h−1Improve weight loss, shortening of the colon, increased DAI score, histological abnormalitiesReduces colonic inflammation and intestinal epithelial cell apoptosis by inhibiting the p38 MAPK/NF-κB signaling pathway(127)
Zhang et al, 2021Rat model of intestinal I/R injury2.5 µg × kg−1 × h−1 and 5 µg × kg−1 × h−1Protection against experimentally induced intestinal I/R injurySuppresses apoptosis of EGCs through SIRT3-mediated PINK1/HDAC3/p53 pathway(128)
Dong et al, 2022Mice model of intestinal I/R injury5 µg·kg−1 × h−1Alleviate intestinal I/R injuryReverses I/R-induced bacterial abnormalities(129)
Xu et al, 2021Rat model of intestinal I/R injury5 µg × kg−1 × h−1Protection against lung injury following intestinal ischemia-reperfusion Ca2+-regulating alpha 2AR/Caveolin-1/ p38MAPK/NF-kappa-B axis(130)
Chang et al, 2020LPS-induced intestinal injury in mice80 µg × kg−1Inhibits gastrointestinal motility disorderMarkedly inhibits gastric emptying, small intestinal transit, colonic transit, gastrointestinal transit and the whole gut transit(131)

[i] DEX, dexmedetomidine; LPS, lipopolysaccharide; TLR4, Toll-like receptor 4; CLP, cecal ligation and puncture; OGD/R, oxygen-glucose deprivation/reoxygenation; DAI, disease activity index; p38MAPK, p38 mitogen-activated protein kinase; NF-κB, nuclear factor-kappa B; EGCs, enteric glial cells; MLCK, Myosin light chain kinase; p-MLC, phosphorylated myosin light chain; MyD88, myeloid differentiation factor-88; SIRT3, Sirtuin-3; PINK1, PTEN-induced putative kinase 1; HDAC3, histone deacetylase 3.

Brain injury

During sepsis, various inflammatory mediators and endotoxins damage capillary endothelial cells, resulting in leakage, inflammatory damage and tight junction dysfunction. This compromises the blood-brain barrier, permitting systemic inflammation and neurotoxic mediators to infiltrate the brain, leading to sepsis-associated brain injury (Fig. 7) (132). Following central nervous system damage, microglia are activated, proliferate rapidly, polarize, migrate to the injury site, and secrete proinflammatory factors such as IL-1β and TNF-α (133), which further degrade the blood-brain barrier, perpetuating microglial activation in a vicious cycle that exacerbates neuronal damage (Fig. 7). Mitigation of microglial activation is crucial for ameliorating sepsis-induced brain injury. Research indicates that DEX attenuates neuroinflammation in LPS-stimulated BV2 microglia by upregulating miR-340 (134) and modulates neuroinflammation and microglial activation through the circ-Shank3/miR-140-3p/TLR4 axis (135). Additionally, DEX inhibits the TLR4/MyD88/NF-κB signaling pathway, reducing the expression of inflammatory factors, such as TNF-α, thereby protecting against ischemia-reperfusion injury (126). By inhibiting MAPK and NF-κB activation, DEX decreases TNF-α and IL-1β levels, shielding brain tissue from inflammatory damage (136). It also enhances microglial M2 polarization by inhibiting ERK1/2 phosphorylation, exerting anti-inflammatory effects in BV2 cells (137). Consequently, DEX mitigates sepsis-induced brain injury through multiple mechanisms. Current research on the protective mechanisms of DEX against sepsis-induced injury has focused primarily on animal and cell studies. Extensive clinical research is necessary to validate these findings and establish a theoretical foundation for the clinical treatment of sepsis-induced injury.

Figure 7

Mechanisms of sepsis-induced brain injury. Endotoxins enter the brain by damaging the blood-brain barrier, activating microglia and leading to neuroinflammation. During sepsis, various inflammatory mediators and endotoxins damage capillary endothelial cells, resulting in leakage and inflammatory damage. Neuroinflammation can lead to oxidative stress in nerve cells, mitochondrial dysfunction, and neuronal apoptosis. Neuroinflammation can increase glutamate production and increase calcium influx into nerve cells, leading to mitochondrial dysfunction and neuronal apoptosis. LPS, lipopolysaccharide; ROS, reactive oxygen species.

Modulating the proinflammatory/anti-inflammatory imbalance

Sepsis involves hyperinflammation and acquired immunosuppression (Fig. 8). Multiple studies (138-140) have indicated immune dysregulation during sepsis. Initially, infection triggers an immune response, leading to the extensive production of inflammatory factors and consequent immune stress, exacerbating organ injury. As sepsis progresses, there is a decline in lymphocyte subsets, further intensifying inflammation. This period is characterized by concurrent immunosuppression and inflammation, which mutually exacerbate sepsis. Thus, immune regulation is pivotal in sepsis treatment. Research on DEX, a novel sedative in sepsis, is expanding, although studies on its impact on immune dysfunction are limited. Wang et al's meta-analysis (52) demonstrated that intraoperative DEX infusion is correlated with i) reduced plasma levels of epinephrine, norepinephrine and cortisol; ii) increased NK cells, B cells and CD4 cells; decreased CD8+ T cells; iii) elevated CD4+CD8+ and Th1:Th2 ratios; iv) reduced TNF-α and IL-6; and increased IL-10. The Th1/Th2 helper T-cell 2 (Th2) and helper T-cell 17 Th17/regulatory T-cell (Treg) ratios reflect immune response dynamics. In patients under surgical and anesthesia stress, DEX elevates the IFN-γ to IL-4 and IL-17 to IL-10 ratios, shifting the Th1/Th2 and Th17/Treg cytokine balance toward Th1 and Th17 in an alcohol-dependent manner and resulting in immunomodulatory effects (141). DEX effectively increases CD3+, CD4+ and NK cells and CD4+/CD8+ levels in patients undergoing radical mastectomy for breast cancer, maintaining cellular immune function homeostasis (142). These studies reveal that DEX possesses anti-inflammatory properties and enhances cellular immunity, making it suitable for septic patients with immunosuppression.

Figure 8

Impact of pathogenic microorganisms on inflammation and immune suppression. During sepsis, endotoxins produced by pathogenic microorganisms stimulate neutrophils, dendritic cells and lymphocytes, producing large amounts of inflammatory cytokines and complement, leading to sepsis, mitochondrial dysfunction and activation of the coagulation system. Moreover, inflammatory cytokines can induce the apoptosis of immune cells, leading to immune suppression. PAMPS, pathogen-associated molecular patterns; DAMPs, damage-associated molecular patterns.

Maintaining hemodynamics

DEX inhibits norepinephrine neuron activity in the locus coeruleus through negative feedback, suppresses sympathetic nerve excitation, and lowers blood catecholamine levels. This stabilizes the intraoperative circulatory status and minimizes the hemodynamic fluctuations induced by surgical trauma and painful stimulation (143,144). Norepinephrine is the preferred vasopressor for restoring mean arterial pressure in septic patients. Refractory septic shock often necessitates higher doses of norepinephrine to maintain blood pressure. However, in sepsis-related AKI, norepinephrine-induced blood pressure restoration exacerbates renal medullary hypoperfusion and hypoxia (145). DEX enhances responsiveness to exogenous vasopressors (146). The results of the present study are shown in Table II (11,117,147-151). DEX significantly reduced catecholamine requirements, halved the required norepinephrine dose, prevented further renal medullary hypoperfusion and hypoxia, and progressively improved creatinine CL, potentially through an enhanced vascular response to catecholamines and angiotensin II. Thus, DEX reduces the norepinephrine dosage in septic patients and is suitable for anesthesia and sedation in severe sepsis and septic shock patients. Nonetheless, large, randomized trials are needed to confirm whether DEX consistently reduces the norepinephrine dosage in septic patients.

Table II

Studies on the effects of DEX on sepsis-induced hemodynamics.

Table II

Studies on the effects of DEX on sepsis-induced hemodynamics.

First author, yearObject of studyDosage of DEXEffectRegulatory mechanism of the hemodynamics(Refs.)
Morelli et al, 2019Patients with septic shock0.7 µg/kg x hSignificantly reducing catecholamine. requirementsInhibits norepinephrine neuron activity in the locus coeruleus.(147)
Lankadeva et al, 2019Sheep model of sepsis-related acute kidney injury0.5 µg/kg x hRestore mean arterial pressure to pre-illness levels, halved the required norepinephrine dose.Enhanced vascular response to catecholamines and angiotensin II.(148)
Taniguchi et al, 2004Endotoxemic rats0.5 µg/kg x hImproving endotoxin induced. hypotensionInhibitory effect on inflammatory response during endotoxemia.(117)
Yao et al, 2021Patients with septic shock0.7 µg/kg x hHas a smaller effect on hemodynamics in patients with septic shock than midazolam.Reduce plasma catecholamine levels in patients with septic shock.(149)
Carnicelli et al, 2022Model of septic shock pigs0.5 µg/kg x hMaintain hemodynamic stability.Not affecting the early metabolic, and inflammatory disorders induced by septic shock.(11)
Moore et al, 2022Patients who underwent rhytidectomy0.7 µg/kg x hResulted in a significant reduction and maintenance of blood pressure from onset of anesthesia.Suppresses sympathetic nerve excitation and lowers blood catecholamine levels.(150)
Hazrati et al, 2022Patients with hip fracture surgery.0.7 µg/kg x hMaintain hemodynamic stability.Reduces the amount of intraoperative bleeding without causing any significant hemodynamic disturbances.(151)

[i] DEX, dexmedetomidine.

Regulating endothelial cells

During sepsis, the activation and dysfunction of vascular endothelial cells result in leukocyte chemotaxis, inflammatory outbreaks, microcirculatory disturbances and altered vascular permeability (Fig. 9) (152). Endothelial cell damage leads to intermittent or complete cessation of capillary blood flow, reducing capillary density and causing nonuniform organ perfusion (153). This endothelial damage-induced microcirculatory disorder results in tissue hypoxia and abnormal cellular oxygenation despite adequate organ blood supply, precipitating organ dysfunction (154). The results of the present study are shown in Table III (87,155-160). Dex pretreatment effectively decreased EC inflammation, apoptosis and hypoxia/reoxygenation injury by inhibiting the DUSP1/MAPK/NF-κB, HMGB1/TLR4/NF-κB, MAPK and angiopoietin II signaling pathways. These studies indicate that DEX protects endothelial barrier function; mitigates sepsis-induced inflammation, microcirculatory disturbances and vascular permeability; and exerts protective effects on vital organs such as the brain, lungs, heart and kidneys during sepsis.

Figure 9

Mechanism by which sepsis induces endothelial cell dysfunction in multiorgan dysfunction. Sepsis can induce the activation and dysfunction of vascular endothelial cells, resulting in leukocyte chemotaxis, inflammatory outbreaks, microcirculatory disturbances and altered vascular permeability.

Table III

Study of the protective mechanism of DEX against sepsis-induced injury to endothelial cells.

Table III

Study of the protective mechanism of DEX against sepsis-induced injury to endothelial cells.

First author, yearObject of studyDosage of DexEffectRegulatory mechanism of the alleviating endothelial cells injury(Refs.)
Dai et al, 2023Angiotensin II-induced endothelial cell100 ng/mlAttenuates Ang II-induced EC dysfunctionInactivating HMGB1/TLR4/NF-κB signaling. pathway(155)
Newton et al, 2002 Statin-statin-induced apoptosis of vascular endothelial cells1 µMBlocks apoptosis of vascular endothelial cellsInhibit serum deprivation, tumor necrosis factor-alpha, oxidants, DNA damage and mitochondrial disruption.(156)
Chai et al, 2020Monocyte-vascular endothelial cells0.1 and 1 nMReduce monocyte-endothelial adherenceInhibit Cx43 alternation regulating the activation of MAPK signaling pathways.(157)
Dong et al, 2020H/R induced mouse pulmonary vascular endothelial cells0.1 nMReduce mouse pulmonary vascular endothelial cells apoptosisIncreased cell viability, reduced apoptotic ratio and downregulated expression levels of Cleaved Caspase-3 and Cleaved Caspase-9 in H/R induced MPVECs(158)
Han et al, 2022LPS induced MPMVECs0.1 nMAlleviate LPS-induced MPMVECs injuryRegulate DUSP1/MAPK/NF-κB axis by inhibiting miR-152-3p(159)
Zhao et al, 2021 Hypoxia/reoxygenation-induced brain endothelial cells0.1 nMAlleviated hypoxia/reoxygenation-induced brain endothelial cells injuryOGD/alleviated Hypoxia/reoxygenation-induced inflammatory injury and permeability in brain endothelial cells mediated by sigma-1 receptor(160)
Zhang et al, 2021CLP-induced rat model of sepsis10 µg/kgProtective effect against CLP-induced endothelial injuryDecreasing angiopoietin 2 and increasing vascular endothelial cadherin levels(87)

[i] DEX, dexmedetomidine; CLP, cecal ligation and puncture; EC, endothelial cell; HMGB1, high mobility group box-1 protein; TLR4, Toll-like receptor 4; NF-κB, nuclear factor kappa B; LPS, lipopolysaccharide; MPVECs, pulmonary microvascular endothelial cells; MAPK, mitogen-activated protein kinase; DUSP1, dual-specificity phosphatase 1.

Reducing postoperative arrhythmias

DEX mitigates postoperative arrhythmias through its antisympathetic effects. Perioperative administration of DEX [with or without a loading dose of 0.2-1.5 µg/kg or an intraoperative maintenance dose of 0.2-1.5 µg/(kg x h)] has been shown to decrease the incidence of atrial fibrillation following cardiac surgery (161), although it may increase the risk of bradycardia (162). A previous large multicenter randomized controlled trial indicated that a lower dose of DEX [0.1-0.4 µg/(kg x h)] as a continuous infusion for 24 h from the induction of anesthesia did not reduce atrial arrhythmias post-cardiac surgery (163). The efficacy of DEX in managing postoperative atrial arrhythmias in septic patients remains controversial and warrants further investigation.

Alleviating brain injury related to general anesthetics

Neurotoxicity associated with general anesthetic drugs is a significant concern. Animal studies have demonstrated that exposure to general anesthetics can induce structural and functional changes in the developing brain, leading to the apoptosis of neurons and glial cells (164,165). The results of the present study are shown in Table IV (166-176). DEX reduced the serum levels of IL-6, TNF-α, MDA and S100β neuron-specific enolase, whereas SOD levels significantly increased 24 h post-surgery, decreased the incidence of postoperative delirium from 23-9%, reduced postoperative agitation within 3 days, inhibited caspase-3 expression in certain cortical and subcortical regions, and exhibited notable neuroprotective effects. However, higher doses of DEX (5 µg/kg) increased mortality rates in rats. Thus, the use of DEX during anesthesia in septic patients may mitigate brain injury caused by sepsis and anesthetic drugs.

Table IV

Study on the protective mechanism of DEX against brain injury related to general anesthetics.

Table IV

Study on the protective mechanism of DEX against brain injury related to general anesthetics.

First author, yearObject of studyDosage of DEXBrain protective effectRegulatory mechanism of the alleviating brain injury(Refs.)
Luo et al, 2016Patients undergoing craniotomy1 µg/kg before anesthesia induction, maintenance dose of 0.4 µg/kg x hAlleviating brain injury by inhibiting inflammationReduces serum levels of IL-6, TNF-α, malondialdehyde, and S100β neuron-specific enolase, while superoxide dismutase levels significantly increased(166)
Sanches et al, 2022Cats subjected to ovariohysterectomy1-5 µg/kg x hAlleviating brain injury by reducing propofol consumption and quicker recovery from anesthesiaReduces propofol consumption and improves cardiorespiratory stability and intraoperative analgesia, while promoting a improved and quicker recovery from anesthesia(167)
Liu et al, 2022Mice undergoing Intracerebral hemorrhage25-100 µg/kgAlleviating brain injury by reducing the neuro-logical damage, brain edema, the lesion volume and pathological damageReducing damage induced by ferroptosis after ICH by regulating iron metabolism, amino acid metabolism and lipid peroxidation processes(168)
Perez-Zoghbi et al, 2017sevoflurane-induced neonatal rats1-50 µg/kgAffording the highest protection in the thalamus (84% reduction) and lowest in the hippocampus and cortical areas (50% reduction)Decreases sevoflurane anesthesia-induced widespread apoptosis(169)
Goyagi, 2019Sevoflurane-induced neonatal rats6.6-50 µg/kgImproving long-term cognitive function and ameliorate the neuronal degenerationSuppresses neuronal apoptosis(170)
Sun et al, 2021Sevoflurane-induced mice10 µg/kgImproved sevoflurane-induced cognitive impairmentInhibits the sevoflurane-induced Tau phosphorylation via activation of alpha-2 adrenergic receptor(171)
Chai et al, 2024Ketamine-induced mice0.1 µMImproved ketamine-anesthesia-induced the neurotoxicity (reduced motor coordination in mice)Mitigated ketamine-induced apoptosis in hippocampal neurons.(172)
Lv et al, 2020Ethanol-neonatal miceProtects against EtOH-mediated inhibition of hippocampal neurogenesisReverses EtOH-induced neuroinflammation via repressing microglia activation and the expression of inflammatory cytokines(173)
Lee et al, 2021Sevoflurane-induced neonatal rats2.5-50 µg/kg/hReduce the anesthetic dosage of Sevofluran and improve the neurotoxicity induced by Sevofluran.Inhibit programmed cell death in several developing brain regions.(174)
Fu et al, 2024Patients undergoing craniocerebral surgery0.5 µg/kg/henhance postoperative cognitive functionMitigates oxidative stress(175)
Shan et al, 2018Sevoflurane-induced rats20 µg/kgImproved sevoflurane-anesthesia-induced the neurotoxicity (including apoptosis and axonal injury, long-term learning and memory dysfunction in the offspring rats)Inhibits neuronal apoptosis by activating the BMP/SMAD signaling pathway(176)

[i] DEX, dexmedetomidine; ICH, intracerebral hemorrhage; EtOH, ethanol; BMPs, bone morphogenetic proteins; Smads, small mothers against decapentaplegic; IL-6, interleukin 6; TNF-α, tumor necrosis factor-α.

Improving coagulation disorders

Pathogenic microorganisms invade the body, triggering immune cells to release proinflammatory mediators that induce tissue factor expression (177). Concurrently, activated coagulation factors engage protein-activated receptors on cell membranes, activating NF-κB and exacerbating inflammation, leading to systemic inflammatory response syndrome, organ damage and potentially MODS (178). The antisympathetic effects of DEX mitigate inflammation, alleviating the hypercoagulable state. Additionally, DEX promotes vasodilation, accelerates blood circulation, reduces red blood cell aggregation, reverses thrombosis, and mitigates organ damage caused by coagulation. Thus, DEX diminishes sepsis-induced inflammation, enhances coagulation function in septic patients, dilates blood vessels, and inhibits the progression of multiple organ failure induced by sepsis.

Future expectations

Although DEX shows promise in reducing organ damage, randomized controlled trials on its efficacy and safety in sepsis are limited. Future research should further assess the therapeutic potential of DEX in sepsis. At present, the protective mechanism of DEX against sepsis-induced damage to organs such as the brain, lungs, liver and heart remains unclear and requires further research. With further research, new targets and clear mechanisms of DEX in the treatment of sepsis may be discovered, expanding its clinical indications beyond perioperative sedatives and providing evidence-based support for promoting patient recovery and improving prognosis. Although numerous studies have reported the organ-protective effects of DEX, numerous aspects of this effect remain controversial. For example, the clinical evidence on whether DEX can reduce postoperative atrial fibrillation and long-term postoperative neurocognitive disorders is not yet unified and still needs to be validated through multicenter large-scale randomized controlled trials.

Conclusions

Sepsis-induced MODS is a leading cause of mortality among critically ill patients and those outside ICUs, with infections primarily originating from the respiratory, digestive and urinary systems. Respiratory infections constitute ~50% of these cases. Sepsis impairs the respiratory and circulatory systems to varying extents, initially presenting as decreased systemic vascular resistance and increased cardiac workload. The severity of sepsis progression is correlated with the characteristics of the infecting bacteria and the patient's immune status. Effective treatment of sepsis-induced MODS hinges on early administration of broad-spectrum antibiotics, stabilization of blood pressure and organ perfusion, timely surgical intervention at the primary infection site and restoration of organ function. Despite these measures, the incidence and mortality rates of sepsis-related MODS remain high (179).

DEX, a highly selective α2-adrenergic receptor agonist initially used for surgical sedation in intensive care units, exhibits anti-inflammatory, antioxidant and antiapoptotic properties both in vivo and in vitro. Research indicates that DEX modulates immune dysfunction, enhances the immune response, lowers cytokine levels, and mitigates organ inflammation during sepsis. It protects against lung and kidney injury, reduces astrocyte inflammatory necrosis in the nervous system, decreases hepatocyte oxidative stress, and inhibits apoptosis pathways, thereby improving circulatory function and reducing mortality in septic rats.

Availability of data and materials

Not applicable.

Authors' contributions

MWL conceptualized the study, acquired funding, and wrote, reviewed and edited the manuscript. SXD curated data. XYZ conducted investigation and design. QFW provided resources, analyzed and interpretated the data. SLY performed software analysis and validation. XL supervised the study and interpretation of data. NM performed visualization and interpretation of data. All authors read and approved the final the version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the Union Foundation of Yunnan Provincial Science and Technology Department and Kunming Medical University (grant no. 202201AY070001-091) and the National Natural Science Foundation of China (grant no. 81960350).

References

1 

Kumar S, Srivastava VK, Kaushik S, Saxena J and Jyoti A: Free radicals, mitochondrial dysfunction and sepsis-induced organ dysfunction: A mechanistic insight. Curr Pharm Des. 30:161–168. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Lelubre C and Vincent JL: Mechanisms and treatment of organ failure in sepsis. Nat Rev Nephrol. 14:417–427. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Keating GM: Dexmedetomidine: A review of its use for sedation in the intensive care setting. Drugs. 75:1119–1130. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Poon WH, Ling RR, Yang IX, Luo H, Kofidis T, MacLaren G, Tham C, Teoh KLK and Ramanathan K: Dexmedetomidine for adult cardiac surgery: A systematic review, meta-analysis and trial sequential analysis. Anesthesia. 78:371–380. 2023. View Article : Google Scholar

5 

Zhang DF, Su X, Meng ZT, Li HL, Wang DX, Xue-Ying Li, Maze M and Ma D: Impact of dexmedetomidine on long-term outcomes after noncardiac surgery in elderly: 3-Year follow-up of a randomized controlled trial. Ann Surg. 270:356–363. 2019. View Article : Google Scholar

6 

Sun YB, Zhao H, Mu DL, Zhang W, Cui J, Wu L, Alam A, Wang DX and Ma D: Dexmedetomidine inhibits astrocyte pyroptosis and subsequently protects the brain in in vitro and in vivo models of sepsis. Cell Death Dis. 10:1672019. View Article : Google Scholar : PubMed/NCBI

7 

Cui J, Zhao H, Yi B, Zeng J, Lu K and Ma D: Dexmedetomidine attenuates bilirubin-induced lung alveolar epithelial cell death in vitro and in vivo. Crit Care Med. 43:e356–e368. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Mei B, Li J and Zuo Z: Dexmedetomidine attenuates sepsis-associated inflammation and encephalopathy via central α2A adrenoceptor. Brain Behav Immun. 91:296–314. 2021. View Article : Google Scholar

9 

Bo JH, Wang JX, Wang XL, Jiao Y, Jiang M, Chen JL, Hao WY, Chen Q, Li YH, Ma ZL and Zhu GQ: Dexmedetomidine attenuates lipopolysaccharide-induced sympathetic activation and sepsis via suppressing superoxide signaling in paraventricular nucleus. Antioxidants (Basel). 11:23952022. View Article : Google Scholar : PubMed/NCBI

10 

Ma S, Evans RG, Iguchi N, Tare M, Parkington HC, Bellomo R, May CN and Lankadeva YR: Sepsis-induced acute kidney injury: A disease of the microcirculation. Microcirculation. 26:e124832019. View Article : Google Scholar

11 

Carnicelli P, Otsuki DA, Filho AM, Kahvegian MAP, Ida KK, Auler JOC Jr, Rouby JJ and Fantoni DT: Effects of dexmedetomidine on hemodynamic, oxygenation, microcirculation, and inflammatory markers in a porcine model of sepsis. Acta Cir Bras. 37:e3707032022. View Article : Google Scholar : PubMed/NCBI

12 

Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al: Global, regional, and national sepsis incidence and mortality, 1990-2017: Analysis for the global burden of disease study. Lancet. 395:200–211. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Arefian H, Heublein S, Scherag A, Brunkhorst FM, Younis MZ, Moerer O, Fischer D and Hartmann M: Hospital-related cost of sepsis: A systematic review. J Infect. 74:107–117. 2017. View Article : Google Scholar

14 

Zhou J, Tian H, Du X, Xi X, An Y, Duan M, Weng L and Du B; for China Critical Care Clinical Trials Group (CCCCTG): Population-based epidemiology of sepsis in a Subdistrict of Beijing. Crit Care Med. 45:1168–1176. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Zhang L, Lin Y, Zhang Z, Chen Y and Zhong J: Immune regulation and organ damage link adiponectin to sepsis. Front Immunol. 15:14448842024. View Article : Google Scholar : PubMed/NCBI

16 

Caraballo C and Jaimes F: Organ dysfunction in sepsis: An ominous trajectory from infection to death. Yale J Biol Med. 92:629–640. 2019.PubMed/NCBI

17 

Killien EY, Zahlan JM, Lad H, Watson RS, Vavilala MS, Huijsmans RLN and Rivara FP: Epidemiology and outcomes of multiple organ dysfunction syndrome following pediatric trauma. J Trauma Acute Care Surg. 93:829–837. 2022. View Article : Google Scholar : PubMed/NCBI

18 

Fleiss N and Polin RA: Sequential organ failure assessment scores to predict outcomes: From adults to neonates. Curr Opin Pediatr. 35:218–222. 2023. View Article : Google Scholar

19 

Nair AS, Saifuddin MS, Naik V and Rayani BK: Dexmedetomidine in cancer surgeries: Present status and consequences with its use. Indian J Cancer. 57:234–238. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Weerink MAS, Struys MMRF, Hannivoort LN, Barends CRM, Absalom AR and Colin P: Clinical pharmacokinetics and pharmacodynamics of dexmedetomidine. Clin Pharmacokinet. 56:893–913. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Behera BK, Misra S, Jena SS and Mohanty CR: The effect of perioperative dexmedetomidine on postoperative bowel function recovery in adult patients receiving general anesthesia. Minerva Anestesiol. 88:51–61. 2022. View Article : Google Scholar

22 

Benet LZ and Hoener BA: Changes in plasma protein binding have little clinical relevance. Clin Pharmacol Ther. 71:115–121. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Gagnon DJ, Riker RR, Glisic EK, Kelner A, Perrey HM and Fraser GL: Transition from dexmedetomidine to enteral clonidine for ICU sedation: An observational pilot study. Pharmacotherapy. 35:251–259. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Sleigh JW, Vacas S, Flexman AM and Talke PO: Electroencephalographic arousal patterns under dexmedetomidine sedation. Anesth Analg. 127:951–959. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Ding J, Chen Y and Gao Y: Effect of propofol, midazolam and dexmedetomidine on ICU patients with sepsis and on arterial blood gas. Exp Ther Med. 18:4340–4346. 2019.PubMed/NCBI

26 

Kawazoe Y, Miyamoto K, Morimoto T, Yamamoto T, Fuke A, Hashimoto A, Koami H, Beppu S, Katayama Y, Itoh M, et al: Effect of dexmedetomidine on mortality and ventilator-free days in patients requiring mechanical ventilation with sepsis: A randomized clinical trial. JAMA. 317:1321–1328. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Miller JW, Balyan R, Dong M, Mahmoud M, Lam JE, Pratap JN, Paquin JR, Li BL, Spaeth JP, Vinks A and Loepke AW: Does intranasal dexmedetomidine provide adequate plasma concentrations for sedation in children: A pharmacokinetic study. Br J Anaesth. 120:1056–1065. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Yadav S and Ramdas E: Intranasal dexmedetomidine versus oral midazolam for premedication in pediatric anesthesia: A prospective double-blinded randomized controlled study. Anesth Analg. 123(3S_Suppl): S3552016. View Article : Google Scholar

29 

Murphy KS, Golden JC and Tampi RR: Dexmedetomidine for agitation in dementia: Current data and future direction. J Am Geriatr Soc. 73:552–557. 2025. View Article : Google Scholar

30 

Scheckenbach V and Fideler F: Optimizing pediatric sedation: Evaluating remimazolam and dexmedetomidine for safety and efficacy in clinical practice. Paediatr Drugs. 27:181–189. 2025. View Article : Google Scholar :

31 

Buckley MS, Smithburger PL, Wong A, Fraser GL, Reade MC, Klein-Fedyshin M, Ardiles T and Kane-Gill SL: Dexmedetomidine for facilitating mechanical ventilation extubation in difficult-to-wean ICU patients: Systematic review and meta-analysis of clinical trials. J Intensive Care Med. 36:925–936. 2021. View Article : Google Scholar

32 

Khan A, Sinha R, Kumar KR, Velpandian T, Maitra S and Ray BR: Comparison of plasma concentration and sedative effect of sublingual and intranasal dexmedetomidine in children: A double-blind randomized controlled study. Acta Anesthesiol Scand. 69:e145832025. View Article : Google Scholar

33 

Portelli K, Kandraju H, Ryu M and Shah PS: Efficacy and safety of dexmedetomidine for analgesia and sedation in neonates: A systematic review. J Perinatol. 44:164–172. 2024. View Article : Google Scholar

34 

Lim JY, Ker CJ, Lai NM, Romantsik O, Fiander M and Tan K: Dexmedetomidine for analgesia and sedation in newborn infants receiving mechanical ventilation. Cochrane Database Syst Rev. 5:CD0123612024.PubMed/NCBI

35 

Kayki G, Yalcin N, Celik HT and Yigit S: Dexmedetomidine in neonates: Utilisation trends and safety profile over time in a neonatal intensive care unit. BMJ Paediatr Open. 9:e0030042025. View Article : Google Scholar : PubMed/NCBI

36 

Ji SJ, Qian ZH, Hu PY and Chen FY: Age-dependent changes in skeletal muscle mass and visceral fat area in a Chinese Population. Curr Med Sci. 43:838–844. 2023. View Article : Google Scholar : PubMed/NCBI

37 

Bucław M, Majewska D, Szczerbińska D and Ligocki M: The influence of age and gender on emu (Dromaius novaehollandiae) fat. Sci Rep. 10:110822020. View Article : Google Scholar

38 

Degrassi I, Leonardi I, Di Profio E, Montanari C, Zuccotti G and Verduci E: Fat-Soluble vitamins deficiency in pediatric cholestasis: A scoping review. Nutrients. 15:24912023. View Article : Google Scholar : PubMed/NCBI

39 

Huang Y, Feng J, Xu J, Dong L, Su W, Li B, Witwer KW and Zheng L: Associations of age and sex with characteristics of extracellular vesicles and protein-enriched fractions of blood plasma. Aging Cell. 24:e143562025. View Article : Google Scholar

40 

Peixoto TC, Quitete FT, Teixeira AVS, Martins BC, Soares RA, Atella GC, Bertasso IM, Lisboa PC, Resende AC, Mucci DB, et al: Palm and interesterified palm oil-enhanced brown fat whitening contributes to metabolic dysfunction in C57BL/6J mice. Nutr Res. 133:94–107. 2025. View Article : Google Scholar

41 

Zhao S, Zhou R, Zhong Q and Zhang M: Effect of age and ICU types on mortality in invasive mechanically ventilated patients with sepsis receiving dexmedetomidine: A retrospective cohort study with propensity score matching. Front Pharmacol. 15:13443272024. View Article : Google Scholar : PubMed/NCBI

42 

Venn RM, Karol MD and Grounds RM: Pharmacokinetics of dexmedetomidine infusions for sedation of postoperative patients requiring intensive caret. Br J Anaesth. 88:669–675. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Hart CG, Voelz BE, Brockus KE and Lemley CO: Hepatic steroid inactivating enzymes, hepatic portal blood flow and corpus luteum blood perfusion in cattle. Reprod Domest Anim. 53:751–758. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Lin L, Guo X, Zhang MZ, Qu CJ, Sun Y and Bai J: Pharmacokinetics of dexmedetomidine in Chinese post-surgical intensive care unit patients. Acta Anesthesiol Scand. 55:359–367. 2011. View Article : Google Scholar

45 

Iirola T, Laitio R, Kentala E, Aantaa R, Kurvinen JP, Scheinin M and Olkkola KT: Highly variable pharmacokinetics of dexmedetomidine during intensive care: A case report. J Med Case Rep. 25:732010. View Article : Google Scholar

46 

Yang L, Zhu L, Qi B, Zhang Y, Ni C, Zhang Y, Shi X, Xia Q, Masters J, Ma D and Yu W: Dexmedetomidine use during orthotopic liver transplantation surgery on early allograft dysfunction: A randomized controlled trial. Int J Surg. 110:5518–5526. 2024. View Article : Google Scholar : PubMed/NCBI

47 

Ustun YB, Koksal E, Turunc E, Komurcu O, Dost B, Ozsay O and Karabulut K: Early extubation after liver transplantation: Is dexmedetomidine a good option?: A retrospective cohort study. Int J Clin Pract. 75:e146292021. View Article : Google Scholar : PubMed/NCBI

48 

Hu H, An S, Sha T, Wu F, Jin Y, Li L, Zeng Z, Wu J and Chen Z: Association between dexmedetomidine administration and outcomes in critically ill patients with sepsis-associated acute kidney injury. J Clin Anesth. 83:1109602022. View Article : Google Scholar : PubMed/NCBI

49 

Evans RG, Iguchi N, Cochrane AD, Marino B, Hood SG, Bellomo R, McCall PR, May CN and Lankadeva YR: Renal hemodynamics and oxygenation during experimental cardiopulmonary bypass in sheep under total intravenous anesthesia. Am J Physiol Regul Integr Comp Physiol. 318:R206–R213. 2020. View Article : Google Scholar :

50 

Neumann MM, Davio MB, Macknet MR and Applegate RL II: Dexmedetomidine for awake fiberoptic intubation in a parturient with spinal muscular atrophy type III for cesarean delivery. Int J Obstet Anesth. 18:403–407. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Aghamiri SM, Samimi AS, Hajian M, Samimi AM and Oroumieh A: Effect of xylazine, detomidine, medetomidine and dexmedetomidine during laparoscopic SCNT embryo transfer on pregnancy rate and some physiological variables in goats. BMC Vet Res. 18:982022. View Article : Google Scholar : PubMed/NCBI

52 

Wang K, Wu M, Xu J, Wu C, Zhang B, Wang G and Ma D: Effects of dexmedetomidine on perioperative stress, inflammation, and immune function: Systematic review and meta-analysis. Br J Anaesth. 123:777–794. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Chen M, Li X and Mu G: Myocardial protective and anti-inflammatory effects of dexmedetomidine in patients undergoing cardiovascular surgery with cardiopulmonary bypass: A systematic review and meta-analysis. J Anesth. 36:5–16. 2022. View Article : Google Scholar

54 

Ma J, Chen Q, Li J, Zhao H, Mi E, Chen Y, Yi B, Ning J, Ma D, Lu K and Gu J: Dexmedetomidine-mediated prevention of renal ischemia-reperfusion injury depends in part on cholinergic anti-inflammatory mechanisms. Anesth Analg. 130:1054–1062. 2020. View Article : Google Scholar

55 

Han QQ, Li XY and Wang YX: Dexmedetomidine attenuates lipopolysaccharide-induced inflammation through macrophageal IL-10 expression following α7 nAchR activation. Int Immunopharmacol. 109:1089202022. View Article : Google Scholar

56 

Li J, Chen Q, He X, Alam A, Ning J, Yi B, Lu K and Gu J: Dexmedetomidine attenuates lung apoptosis induced by renal ischemia-reperfusion injury through α2AR/PI3K/Akt pathway. J Transl Med. 16:782018. View Article : Google Scholar

57 

Wang K and Zhu Y: Dexmedetomidine protects against oxygen-glucose deprivation/reoxygenation injury-induced apoptosis via the p38 MAPK/ERK signalling pathway. J Int Med Res. 46:675–686. 2018. View Article : Google Scholar :

58 

Zhu Z, Ling X, Zhou H, Zhang C and Yan W: Dexmedetomidine attenuates cellular injury and apoptosis in H9c2 cardiomyocytes by regulating p-38MAPK and endoplasmic reticulum stress. Drug Des Dev Ther. 14:4231–4243. 2020. View Article : Google Scholar

59 

Deng X, Ye F, Zeng L, Luo W, Tu S, Wang X and Zhang Z: Dexmedetomidine mitigates myocardial ischemia/reperfusion-induced mitochondrial apoptosis through targeting lncRNA HCP5. Am J Chin Med. 50:1529–1551. 2022. View Article : Google Scholar : PubMed/NCBI

60 

Wei Q, Chen J, Xiao F, Tu Y, Zhong Y and Xie Y: High-dose dexmedetomidine promotes apoptosis in fetal rat hippocampal neurons. Drug Des Dev Ther. 15:2433–2444. 2021. View Article : Google Scholar

61 

Zhang Z, Mu X and Zhou X: Dexmedetomidine alleviates inflammatory response and oxidative stress injury of vascular smooth muscle cell via α2AR/GSK-3β/MKP-1/NRF2 axis in intracranial aneurysm. BMC Pharmacol Toxico. 23:812022. View Article : Google Scholar

62 

Choi HI, Kim HJ, Park JS, Kim IJ, Bae EH, Ma SK and Kim SW: PGC-1α attenuates hydrogen peroxide-induced apoptotic cell death by upregulating Nrf-2 via GSK3β inactivation mediated by activated p38 in HK-2 cells. Sci Rep. 7:43192017. View Article : Google Scholar

63 

Zhao Y, Kong GY, Pei WM, Zhou B, Zhang QQ and Pan BB: Dexmedetomidine alleviates hepatic injury via the inhibition of oxidative stress and activation of the Nrf2/HO-1 signaling pathway. Eur Cytokine Netw. 30:88–97. 2019. View Article : Google Scholar

64 

Chen J, Shen N, Duan X and Guo Y: An investigation of the mechanism of dexmedetomidine in improving postoperative cognitive dysfunction from the perspectives of alleviating neuronal mitochondrial membrane oxidative stress and electrophysiological dysfunction. Exp Ther Med. 15:2037–2043. 2018.PubMed/NCBI

65 

Yu T, Liu D, Gao M, Yang P, Zhang M, Song F, Zhang X and Liu Y: Dexmedetomidine prevents septic myocardial dysfunction in rats via activation of α7nAChR and PI3K/Akt-mediated autophagy. Biomed Pharmacother. 120:1092312019. View Article : Google Scholar

66 

Lempiäinen J, Finckenberg P, Mervaala EE, Storvik M, Kaivola J, Lindstedt K, Levijoki J and Mervaala EM: Dexmedetomidine preconditioning ameliorates kidney ischemia-reperfusion injury. Pharmacol Res Perspe. 2:e000452014. View Article : Google Scholar

67 

Li Y, Qu M, Xing F, Li H, Cheng D, Xing N and Zhang W: The protective mechanism of dexmedetomidine in regulating Atg14L-Beclin1-Vps34 complex against myocardial ischemia-reperfusion injury. J Cardiovasc Transl Res. 14:1063–1074. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Zhu Y, Li S, Liu J, Wen Q, Yu J, Yu L and Xie K: Role of JNK signaling pathway in dexmedetomidine post-conditioning-induced reduction of the inflammatory response and autophagy effect of focal cerebral ischemia reperfusion injury in rats. Inflammation. 42:2181–2191. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Qin W, Zhang X, Yang S, Li Y, Yuan J, Yang L, Li S and Hu W: Risk factors for multiple organ dysfunction syndrome in severe stroke patients. PLoS One. 11:e01671892016. View Article : Google Scholar : PubMed/NCBI

70 

van der Poll T, Shankar-Hari M and Wiersinga WJ: The immunology of sepsis. Immunity. 54:2450–2464. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Gordon S: Phagocytosis: An immunobiologic process. Immunity. 44:463–475. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Kumar V: Immunometabolism: Another road to sepsis and its therapeutic targeting. Inflammation. 42:765–788. 2019. View Article : Google Scholar

73 

Hoesel LM, Neff TA, Neff SB, Younger JG, Olle EW, Gao H, Pianko MJ, Bernacki KD, Sarma JV and Ward PA: Harmful and protective roles of neutrophils in sepsis. Shock. 24:40–47. 2005. View Article : Google Scholar : PubMed/NCBI

74 

Lu J, Liu J and Li A: Roles of neutrophil reactive oxygen species (ROS) generation in organ function impairment in sepsis. J Zhejiang Univ Sci B. 23:437–450. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Christoffersson G, Vågesjö E, Vandooren J, Lidén M, Massena S, Reinert RB, Brissova M, Powers AC, Opdenakker G and Phillipson M: VEGF-A recruits a proangiogenic MMP-9-delivering neutrophil subset that induces angiogenesis in transplanted hypoxic tissue. Blood. 120:4653–4662. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Bateman RM, Sharpe MD and Ellis CG: Bench-to-bedside review: Microvascular dysfunction in sepsis-hemodynamics, oxygen transport, and nitric oxide. Crit Care. 7:359–373. 2003. View Article : Google Scholar : PubMed/NCBI

77 

Wang Y, Bai C and Wang X: COPD-associated vascular pathology: A future targeting area. Expert Rev Respir Med. 2:297–299. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Fein AM and Calalang-Colucci MG: Acute lung injury and acute respiratory distress syndrome in sepsis and septic shock. Crit Care Clin. 16:289–317. 2000. View Article : Google Scholar : PubMed/NCBI

79 

Chen G, Hou Y, Li X, Pan R and Zhao D: Sepsis-induced acute lung injury in young rats is relieved by calycosin through inactivating the HMGB1/MyD88/NF-κB pathway and NLRP3 inflammasome. Int Immunopharmacol. 96:1076232021. View Article : Google Scholar

80 

Li J, Lu K, Sun F, Tan S, Zhang X, Sheng W, Hao W, Liu M, Lv W and Han W: Panaxydol attenuates ferroptosis against LPS-induced acute lung injury in mice by Keap1-Nrf2/HO-1 pathway. J Transl Med. 19:962021. View Article : Google Scholar : PubMed/NCBI

81 

Hou L, Zhang J, Liu Y, Fang H, Liao L, Wang Z, Yuan J, Wang X, Sun J, Tang B, et al: MitoQ alleviates LPS-mediated acute lung injury through regulating Nrf2/Drp1 pathway. Free Radic Biol Med. 165:219–228. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Wu Y, Liu Y, Huang H, Zhu Y, Zhang Y, Lu F and Zhou C, Huang L, Li X and Zhou C: Dexmedetomidine inhibits inflammatory reaction in lung tissues of septic rats by suppressing TLR4/NF-κB pathway. Mediators Inflamm. 2013:5621542013. View Article : Google Scholar

83 

Cui H and Zhang Q: Dexmedetomidine ameliorates lipopolysaccharide-induced acute lung injury by inhibiting the PI3K/Akt/FoxO1 signaling pathway. J Anesth. 35:394–404. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Fu C, Dai X, Yang Y, Lin M, Cai Y and Cai S: Dexmedetomidine attenuates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress, mitochondrial dysfunction and apoptosis in rats. Mol Med Rep. 15:131–138. 2017. View Article : Google Scholar :

85 

Shi J, Yu T, Song K, Du S, He S, Hu X, Li X, Li H, Dong S, Zhang Y, et al: Dexmedetomidine ameliorates endotoxin-induced acute lung injury in vivo and in vitro by preserving mitochondrial dynamic equilibrium through the HIF-1a/HO-1 signaling pathway. Redox Biol. 41:1019542021. View Article : Google Scholar : PubMed/NCBI

86 

Meng L, Li L, Lu S, Li K, Su Z, Wang Y, Fan X, Li X and Zhao G: The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-κB and PI3K/Akt/mTOR pathways. Mol Immunol. 94:7–17. 2018. View Article : Google Scholar

87 

Zhang P, Peng J, Ren YQ, Zheng H and Yan H: Dexmedetomidine protects against endothelial injury in septic rats induced by cecal ligation and puncture by decreasing angiopoietin 2 and increasing vascular endothelial cadherin levels. Exp Ther Med. 21:1112021. View Article : Google Scholar

88 

Wang R, Xie Y, Qiu J and Chen J: The effects of dexmedetomidine in a rat model of sepsis-induced lung injury are mediated through the adenosine monophosphate-activated protein kinase (AMPK)/silent information regulator 1 (SIRT1) pathway. Med Sci Monit. 26:e9192132020.PubMed/NCBI

89 

Peerapornratana S, Manrique-Caballero CL, Gómez H and Kellum JA: Acute kidney injury from sepsis: Current concepts, epidemiology, pathophysiology, prevention and treatment. Kidney Int. 96:1083–1099. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Manrique-Caballero CL, Del Rio-Pertuz G and Gomez H: Sepsis-associated acute kidney injury. Crit Care Clin. 37:279–301. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Hu X, Zhou W, Wu S, Wang R, Luan Z, Geng X, Xu N, Zhang Z, Ruan Z, Wang Z, et al: Tacrolimus alleviates LPS-induced AKI by inhibiting TLR4/MyD88/NF-κB signalling in mice. J Cell Mol Med. 26:507–514. 2022. View Article : Google Scholar

92 

Huang G, Bao J, Shao X, Zhou W, Wu B, Ni Z and Wang L: Inhibiting pannexin-1 alleviates sepsis-induced acute kidney injury via decreasing NLRP3 inflammasome activation and cell apoptosis. Life Sci. 254:1177912020. View Article : Google Scholar : PubMed/NCBI

93 

Li J, Wang L, Wang B, Zhang Z, Jiang L, Qin Z, Zhao Y and Su B: NOX4 is a potential therapeutic target in septic acute kidney injury by inhibiting mitochondrial dysfunction and inflammation. Theranostics. 13:2863–2878. 2023. View Article : Google Scholar : PubMed/NCBI

94 

Liu W, Yu W, Weng Y, Wang Y and Sheng M: Dexmedetomidine ameliorates the inflammatory immune response in rats with acute kidney damage. Exp Ther Med. 14:3602–3608. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Jin YH, Li ZT, Chen H, Jiang XQ, Zhang YY and Wu F: Effect of dexmedetomidine on kidney injury in sepsis rats through TLR4/MyD88/NF-κB/iNOS signaling pathway. Eur Rev Med Pharmacol Sci. 23:5020–5025. 2019.PubMed/NCBI

96 

Yao Y, Hu X, Feng X, Zhao Y, Song M, Wang C and Fan H: Dexmedetomidine alleviates lipopolysaccharide-induced acute kidney injury by inhibiting the NLRP3 inflammasome activation via regulating the TLR4/NOX4/NF-κB pathway. J Cell Biochem. 120:18509–18523. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Wang Z, Wu J, Hu Z, Luo C, Wang P, Zhang Y and Li H: Dexmedetomidine alleviates lipopolysaccharide-induced acute kidney injury by inhibiting p75NTR-mediated oxidative stress and apoptosis. Oxid Med Cell Longev. 2020:54542102020. View Article : Google Scholar : PubMed/NCBI

98 

Zhao Y, Feng X, Li B, Sha J, Wang C, Yang T, Cui H and Fan H: Dexmedetomidine protects against lipopolysaccharide-induced acute kidney injury by enhancing autophagy through inhibition of the PI3K/AKT/mTOR pathway. Front Pharmacol. 11:1282020. View Article : Google Scholar : PubMed/NCBI

99 

Kim TS and Choi DH: Liver dysfunction in sepsis. Korean J Gastroenterol. 75:182–187. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Cai Y, Zhao D, Pan Y, Chen B, Cao Y, Han S, Lian F, Zhang Y and Yan X: Gallic acid attenuates sepsis-induced liver injury through C/EBPbeta-dependent MAPK signaling pathway. Mol Nutr Food Res. 29:e24001232024. View Article : Google Scholar

101 

Ou Z, Zhong H, Zhang L, Deng M, Zhang W, Wang J, Feng H, Gong J, Miao C and Yi Z: Macrophage membrane-coated nanoparticles alleviate hepatic ischemia-reperfusion injury caused by orthotopic liver transplantation by neutralizing endotoxin. Int J Nanomedicine. 15:4125–4138. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Song H, Zhang X, Zhai R, Liang H, Song G, Yuan Y, Xu Y, Yan Y, Qiu L and Sun T: Metformin attenuated sepsis-associated liver injury and inflammatory response in aged mice. Bioengineered. 13:4598–4609. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Hong KS, Kim NR, Song SH and Hong G: Cycling of dexmedetomidine may prevent delirium after liver transplantation. Transplant Proc. 50:1080–1082. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Tong F, Shen W, Song P, Song J, Hu Y, Liu F, Meng Z and Liu J: Dexmedetomidine attenuates lipopolysaccharide-induced acute liver injury in rats by inhibiting caveolin-1 downstream signaling pathway. Biosci Rep. 41:BSR202042792021. View Article : Google Scholar : PubMed/NCBI

105 

Zi SF, Li JH, Liu L, Deng C, Ao X, Chen DD and Wu SZ: Dexmedetomidine-mediated protection against septic liver injury depends on TLR4/MyD88/NF-κB signaling downregulation partly via cholinergic anti-inflammatory mechanisms. Int Immunopharmacol. 76:1058982019. View Article : Google Scholar

106 

Yu Q, Zou L, Yuan X, Fang F and Xu F: Dexmedetomidine protects against septic liver injury by enhancing autophagy through activation of the AMPK/SIRT1 signaling pathway. Front Pharmacol. 12:6586772021. View Article : Google Scholar : PubMed/NCBI

107 

Sha J, Zhang H, Zhao Y, Feng X, Hu X, Wang C, Song M and Fan H: Dexmedetomidine attenuates lipopolysaccharide-induced liver oxidative stress and cell apoptosis in rats by increasing GSK-3β/MKP-1/Nrf2 pathway activity via the α2 adrenergic receptor. Toxicol Appl Pharmacol. 364:144–152. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Zhou H, Sun J, Zhong W, Pan X, Liu C, Cheng F, Wang P and Rao Z: Dexmedetomidine preconditioning alleviated murine liver ischemia and reperfusion injury by promoting macrophage M2 activation via PPARγ/STAT3 signaling. Int Immunopharmacol. 82:1063632020. View Article : Google Scholar

109 

Frencken JF, Donker DW, Spitoni C, Koster-Brouwer ME, Soliman IW, Ong DSY, Horn J, van der Poll T, van Klei WA, Bonten MJM and Cremer OL: Myocardial injury in patients with sepsis and its association with long-term outcome. Circ Cardiovasc Qual Outcomes. 11:e0040402018. View Article : Google Scholar : PubMed/NCBI

110 

Orde SR, Pulido JN, Masaki M, Gillespie S, Spoon JN, Kane GC and Oh JK: Outcome prediction in sepsis: Speckle tracking echocardiography based assessment of myocardial function. Crit Care. 18:R1492014. View Article : Google Scholar : PubMed/NCBI

111 

Maeder M, Fehr T, Rickli H and Ammann P: Sepsis-associated myocardial dysfunction: Diagnostic and prognostic impact of cardiac troponins and natriuretic peptides. Chest. 129:1349–1366. 2006. View Article : Google Scholar : PubMed/NCBI

112 

Sun F, Xu K, Zhou J, Zhang W, Duan G and Lei M: Allicin protects against LPS-induced cardiomyocyte injury by activating Nrf2-HO-1 and inhibiting NLRP3 pathways. BMC Cardiovasc Disord. 23:4102023. View Article : Google Scholar : PubMed/NCBI

113 

Wang Y, Feng W, Li S, Liu C, Jia L, Wang P, Li L, Du H and Yu W: Oxycodone attenuates lipopolysaccharide-induced myocardial injury by inhibiting inflammation, oxidation and pyroptosis via Nrf2/HO-1 signalling pathway. Clin Exp Pharmacol Physiol. 51:e139102024. View Article : Google Scholar : PubMed/NCBI

114 

Lu C, Liu J, Escames G, Yang Y, Wu X, Liu Q, Chen J, Song Y, Wang Z, Deng C, et al: PIK3CG regulates NLRP3/ GSDMD-mediated pyroptosis in septic myocardial injury. Inflammation. 46:2416–2432. 2023. View Article : Google Scholar : PubMed/NCBI

115 

Bi CF, Liu J, Yang LS and Zhang JF: Research progress on the mechanism of sepsis induced myocardial injury. J Inflamm Res. 15:4275–4290. 2022. View Article : Google Scholar : PubMed/NCBI

116 

Taniguchi T, Kurita A, Kobayashi K, Yamamoto K and Inaba H: Dose- and time-related effects of dexmedetomidine on mortality and inflammatory responses to endotoxin-induced shock in rats. J Anesth. 22:221–228. 2008. View Article : Google Scholar : PubMed/NCBI

117 

Taniguchi T, Kidani Y, Kanakura H, Takemoto Y and Yamamoto K: Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Crit Care Med. 32:1322–1326. 2004. View Article : Google Scholar : PubMed/NCBI

118 

Kong W, Kang K, Gao Y, Liu H, Meng X, Yang S, Yu K and Zhao M: Dexmedetomidine alleviates LPS-induced septic cardiomyopathy via the cholinergic anti-inflammatory pathway in mice. Am J Transl Res. 9:5040–5047. 2017.PubMed/NCBI

119 

Wang C, Yuan W, Hu A, Lin J, Xia Z, Yang CF, Li Y and Zhang Z: Dexmedetomidine alleviated sepsis-induced myocardial ferroptosis and septic heart injury. Mol Med Rep. 22:175–184. 2020. View Article : Google Scholar : PubMed/NCBI

120 

Zhang T, Mei Q, Dai S, Liu Y and Zhu H: Use of dexmedetomidine in patients with sepsis: A systematic review and meta-analysis of randomized-controlled trials. Ann Intensive Care. 12:812022. View Article : Google Scholar : PubMed/NCBI

121 

Pool R, Gomez H and Kellum JA: Mechanisms of organ dysfunction in sepsis. Crit Care Clin. 34:63–80. 2018. View Article : Google Scholar

122 

Haussner F, Chakraborty S, Halbgebauer R and Huber-Lang M: Challenge to the intestinal mucosa during sepsis. Front Immunol. 10:8912019. View Article : Google Scholar : PubMed/NCBI

123 

Rowlands BJ, Soong CV and Gardiner KR: The gastrointestinal tract as a barrier in sepsis. Br Med Bull. 55:196–211. 1999. View Article : Google Scholar

124 

Chen Y, Miao L, Yao Y, Wu W, Wu X, Gong C, Qiu L and Chen J: Dexmedetomidine ameliorate CLP-induced rat intestinal injury via inhibition of inflammation. Mediators Inflamm. 2015:9183612015. View Article : Google Scholar : PubMed/NCBI

125 

Qin C, Jiang Y, Chen X, Bian Y, Wang Y, Xie K and Yu Y: Dexmedetomidine protects against burn-induced intestinal barrier injury via the MLCK/p-MLC signalling pathway. Burns. 47:1576–1585. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Yang J, Wu Y, Xu Y, Jia J, Xi W, Deng H and Tu W: Dexmedetomidine resists intestinal Ischemia-reperfusion injury by inhibiting TLR4/MyD88/NF-κB signaling. J Surg Res. 260:350–358. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Ye X, Xu H and Yuan X: Dexmedetomidine alleviates intestinal barrier dysfunction and inflammatory response in mice via suppressing TLR4/MyD88/NF-κB signaling in an experimental model of ulcerative colitis. Folia Histochem Cytobiol. 60:311–322. 2022. View Article : Google Scholar

128 

Zhang Q, Liu XM, Hu Q, Liu ZR, Liu ZY, Zhang HG, Huang YL, Chen QH, Wang WX and Zhang XK: Dexmedetomidine inhibits mitochondria damage and apoptosis of enteric glial cells in experimental intestinal ischemia/reperfusion injury via SIRT3-dependent PINK1/HDAC3/p53 pathway. J Transl Med. 19:4632021. View Article : Google Scholar : PubMed/NCBI

129 

Dong YH, Hu JJ, Deng F, Chen XD, Li C, Liu KX and Zhao BC: Use of dexmedetomidine to alleviate intestinal ischemia-reperfusion injury via intestinal microbiota modulation in mice. Ann Transl Med. 10:11612022. View Article : Google Scholar : PubMed/NCBI

130 

Xu L, Li T, Chen Q, Liu Z, Chen Y, Hu K and Zhang X: The α2AR/Caveolin-1/p38MAPK/NF-κB axis explains dexmedetomidine protection against lung injury following intestinal ischaemia-reperfusion. J Cell Mol Med. 25:6361–6372. 2021. View Article : Google Scholar : PubMed/NCBI

131 

Chang H, Li S, Li Y, Hu H, Cheng B, Miao J, Gao H, Ma H, Gao Y and Wang Q: Effect of sedation with dexmedetomidine or propofol on gastrointestinal motility inlipopolysaccharide-induced endotoxemic mice. BMC Anesthesiol. 20:2272020. View Article : Google Scholar

132 

Zhao L, Gao Y, Guo S, Lu X, Yu S, Ge ZZ, Zhu HD and Li Y: Sepsis-associated encephalopathy: Insight into injury and pathogenesis. CNS Neurol Disord Drug Targets. 20:112–124. 2021.

133 

Subhramanyam CS, Wang C, Hu Q and Dheen ST: Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin Cell Dev Biol. 94:112–120. 2019. View Article : Google Scholar : PubMed/NCBI

134 

Bao Y, Zhu Y, He G, Ni H, Liu C, Ma L, Zhang L and Shi D: Dexmedetomidine attenuates neuroinflammation in LPS-stimulated BV2 microglia cells through upregulation Of miR-340. Drug Des Devel Ther. 13:3465–3475. 2019. View Article : Google Scholar : PubMed/NCBI

135 

He G, He Y, Ni H, Wang K, Zhu Y and Bao Y: Dexmedetomidine attenuates neuroinflammation and microglia activation in LPS-stimulated BV2 microglia cells through targeting circ-Shank3/miR-140-3p/TLR4 axis. Eur J Histochem. 67:37662023. View Article : Google Scholar : PubMed/NCBI

136 

Lei S, Lu P, Lu Y, Zheng J, Li W, Wang N, Zhang H, Li R, Wang K, Wen J, et al: Dexmedetomidine alleviates neurogenesis damage following neonatal midazolam exposure in rats through JNK and P38 MAPK pathways. ACS Chem Neurosci. 11:579–591. 2020. View Article : Google Scholar : PubMed/NCBI

137 

Sun Z, Lin Y, Li Y, Ren T, Du G, Wang J, Jin X and Yang LC: The effect of dexmedetomidine on inflammatory inhibition and microglial polarization in BV-2 cells. Neurol Res. 40:838–846. 2018. View Article : Google Scholar : PubMed/NCBI

138 

Hamers L, Kox M and Pickkers P: Sepsis-induced immunoparalysis: Mechanisms, markers, and treatment options. Minerva Anestesiol. 81:426–439. 2015.

139 

Martin MD, Badovinac VP and Griffith TS: CD4 T cell responses and the sepsis-induced immunoparalysis state. Front Immunol. 11:13642020. View Article : Google Scholar : PubMed/NCBI

140 

Łysenko L, Leśnik P, Nelke K and Gerber H: Immune disorders in sepsis and their treatment as a significant problem of modern intensive care. Postepy Hig Med Dosw (Online). 71:703–712. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Lee JM, Han HJ, Choi WK, Yoo S, Baek S and Lee J: Immunomodulatory effects of intraoperative dexmedetomidine on T helper 1, T helper 2, T helper 17 and regulatory T cells cytokine levels and their balance: A prospective, randomised, double-blind, dose-response clinical study. BMC Anesthesiol. 18:1642018. View Article : Google Scholar : PubMed/NCBI

142 

Yang XH, Bai Q, LvMM, Fu HG, Dong TL and Zhou Z: Effect of dexmedetomidine on Immune function of patients undergoing radical mastectomy: A double blind and placebo control study. Eur Rev Med Pharmacol Sci. 21:1112–1116. 2017.PubMed/NCBI

143 

Biccard BM, Goga S and de Beurs J: Dexmedetomidine and cardiac protection for non-cardiac surgery: A meta-analysis of randomised controlled trials. Anaesthesia. 63:4–14. 2008. View Article : Google Scholar

144 

Farouk I, Hassan MM, Fetouh AM, Elgayed AEA, Eldin MH and Abdelhamid BM: Analgesic and hemodynamic effects of intravenous infusion of magnesium sulphate versus dexmedetomidine in patients undergoing bilateral inguinal hernial surgeries under spinal anesthesia: A randomized controlled study. Braz J Anesthesiol. 71:489–497. 2021.PubMed/NCBI

145 

Lankadeva YR, Kosaka J, Evans RG, Bailey SR, Bellomo R and May CN: Intrarenal and urinary oxygenation during norepinephrine resuscitationin ovine septic acute kidney injury. Kidney Int. 90:100–108. 2016. View Article : Google Scholar : PubMed/NCBI

146 

Julien C, Oréa V, Quintin L, Piriou V and Barrès C: Renal sympathetic nerve activity and vascular reactivity to phenylephrine after lipopolysaccharide administration in conscious rats. Physiol Rep. 5:e131392017. View Article : Google Scholar : PubMed/NCBI

147 

Morelli A, Sanfilippo F, Arnemann P, Hessler M, Kampmeier TG, D'Egidio A, Orecchioni A, Santonocito C, Frati G, Greco E, et al: The effect of propofol and dexmedetomidine sedation on norepinephrine requirements in septic shock patients: A crossover trial. Crit Care Med. 47:e89–e95. 2019. View Article : Google Scholar

148 

Lankadeva YR, Ma S, Iguchi N, Evans RG, Hood SG, Farmer DGS, Bailey SR, Bellomo R and May CN: Dexmedetomidine reduces norepinephrine requirements and preserves renal oxygenation and function in ovine septic acute kidney injury. Kidney Int. 96:1150–1161. 2019. View Article : Google Scholar : PubMed/NCBI

149 

Yao L, Liu B, He T and Shen F: Comparative study of dexmedetomidine vs. midazolam on plasma catecholamine levels and hemodynamics in patients with septic shock. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 33:1193–1197. 2021.In Chinese. PubMed/NCBI

150 

Moore ACM, Kachare SD, Barber DA, Barrow L and O'Daniel TG: Total intravenous anesthesia with dexmedetomidine for hemodynamic stability and enhanced recovery in facial aesthetic surgery. Aesthet Surg J. 42:NP602–NP610. 2022. View Article : Google Scholar : PubMed/NCBI

151 

Hazrati E, Vosoughi F, Chamanara M and Teymourian H: Effect of dexmedetomidine infusion during hip fracture surgery on hemodynamic parameters and blood loss: A triple-blinded randomized clinical trial. Injury. 53:551–554. 2022. View Article : Google Scholar

152 

Joffre J, Hellman J, Ince C and Ait-Oufella H: Endothelial responses in sepsis. Am J Respir Crit Care Med. 202:361–370. 2020. View Article : Google Scholar : PubMed/NCBI

153 

Joffre J and Hellman J: Oxidative stress and endothelial dysfunction in sepsis and acute inflammation. Antioxid Redox Signal. 35:1291–1307. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Cusack R, Leone M, Rodriguez AH and Martin-Loeches I: Endothelial Damage And the microcirculation in critical illness. Biomedicines. 10:31502022. View Article : Google Scholar : PubMed/NCBI

155 

Dai M, Zhu X, Zeng S, Liu Q, Hu R, Huang L, Wang Y, Deng J and Yu Q: Dexmedetomidine protects cells from angiotensin II-induced smooth muscle cell phenotype switch and endothelial cell dysfunction. Cell Cycle. 22:450–463. 2023. View Article : Google Scholar :

156 

Newton CJ, Ran G, Xie YX, Bilko D, Burgoyne CH, Adams I, Abidia A, McCollum PT and Atkin SL: Statin-induced apoptosis of vascular endothelial cells is blocked by dexamethasone. J Endocrinol. 174:7–16. 2002. View Article : Google Scholar : PubMed/NCBI

157 

Chai Y, Yu R, Liu Y, Wang S, Yuan D and Chen J: Dexmedetomidine attenuates monocyte-endothelial adherence via inhibiting connexin43 on vascular endothelial cells. Mediators Inflamm. 2020:70398542020. View Article : Google Scholar : PubMed/NCBI

158 

Dong W, Yang H, Cheng M, Zhang X, Yin J, Zeng Z and Huang G: Dexmedetomidine alleviates pulmonary ischemia-reperfusion injury through modulating the miR-21-5p/Nr4a1 signaling pathway. Acta Biochim Pol. 67:521–529. 2020.PubMed/NCBI

159 

Han J, Liu X and Wang L: Dexmedetomidine protects against acute lung injury in mice via the DUSP1/MAPK/NF-κB axis by inhibiting miR-152-3p. Pulm Pharmacol Ther. 9:1021312022. View Article : Google Scholar

160 

Zhao Q, Yu S, Ling Y, Hao S and Liu J: The protective effects of dexmedetomidine against hypoxia/reoxygenation-induced inflammatory injury and permeability in brain endothelial cells mediated by sigma-1 receptor. ACS Chem Neurosci. 12:1940–1947. 2021. View Article : Google Scholar : PubMed/NCBI

161 

Zhu Z, Zhou H, Ni Y, Wu C, Zhang C and Ling X: Can dexmedetomidine reduce atrial fibrillation after cardiac surgery? A systematic review and meta-analysis. Drug Des Devel Ther. 12:521–531. 2018. View Article : Google Scholar : PubMed/NCBI

162 

Liu X, Xie G, Zhang K, Song S, Song F, Jin Y and Fang X: Dexmedetomidine vs propofol sedation reduces delirium in patients after cardiac surgery: A meta analysis with trial sequential analysis of randomized controlled trials. J Crit Care. 38:190–196. 2017. View Article : Google Scholar

163 

Turan A, Duncan A, Leung S, Karimi N, Fang J, Mao G, Hargrave J, Gillinov M, Trombetta C, Ayad S, et al: Dexmedetomidine for reduction of atrial fibrillation and delirium after cardiac surgery(DECADE): A randomised placebo-controlled trial. Lancet. 396:177–185. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Hu X, Sun Z, Wang W, Xiao G, Yu Q, Chi L and Liu H: Dexmedetomidine attenuates isoflurane-induced neuroapoptosis through the miR-137/GSK-3β pathway in the developing rat hippocampus. Heliyon. 10:e313722024. View Article : Google Scholar

165 

Wu Q, Shang Y, Shen T, Liu F and Zhang W: Biochanin A protects SH-SY5Y cells against isoflurane-induced neurotoxicity by suppressing oxidative stress and apoptosis. Neurotoxicology. 86:10–18. 2021. View Article : Google Scholar : PubMed/NCBI

166 

Luo X, Zheng X and Huang H: Protective effects of dexmedetomidine on brain function of glioma patients undergoing craniotomy resection and its underlying mechanism. Clin Neurol Neurosurg. 146:105–108. 2016. View Article : Google Scholar : PubMed/NCBI

167 

Sanches MC, Bessi WH, Rusch E, Schaffhausser MB, Cassoli AA, Freitas SH, Gehrcke MI and Carregaro AB: Cardiopulmonary and propofol-sparing effects of dexmedetomidine in total intravenous anesthesia in cats undergoing ovariohysterectomy. J Feline Med Surg. 24:e490–e497. 2022. View Article : Google Scholar : PubMed/NCBI

168 

Liu MJ, Zhao XC, Gong HS, You YQ and Li JY: Dexmedetomidine prevents hemorrhagic brain injury by reducing damage induced by ferroptosis in mice. Neurosci Lett. 788:1368422022. View Article : Google Scholar : PubMed/NCBI

169 

Perez-Zoghbi JF, Zhu W, Grafe MR and Brambrink AM: Dexmedetomidine-mediated neuroprotection against sevoflurane-induced neurotoxicity extends to several brain regions in neonatal rats. Br J Anaesth. 119:506–516. 2017. View Article : Google Scholar : PubMed/NCBI

170 

Goyagi T: Dexmedetomidine reduced sevoflurane-induced neurodegeneration and long-term memory deficits in neonatal rats. Int J Dev Neurosci. 75:19–26. 2019. View Article : Google Scholar : PubMed/NCBI

171 

Sun M, Dong Y, Li M, Zhang Y, Liang F, Zhang J, Soriano SG and Xie Z: Dexmedetomidine and clonidine attenuate sevoflurane-induced tau phosphorylation and cognitive impairment in young mice via α-2 adrenergic receptor. Anesth Analg. 132:878–889. 2021. View Article : Google Scholar :

172 

Chai D, Jiang H and Lv X: The impact of dexmedetomidine on ketamine-induced neurotoxicity and cognitive impairment in young mice. Int J Dev Neurosci. 84:735–744. 2024. View Article : Google Scholar : PubMed/NCBI

173 

Lv K, Yang C, Xiao R, Yang L, Liu T, Zhang R and Fan X: Dexmedetomidine attenuates ethanol-induced inhibition of hippocampal neurogenesis in neonatal mice. Toxicol Appl Pharmacol. 390:1148812020. View Article : Google Scholar : PubMed/NCBI

174 

Lee JR, Joseph B, Hofacer RD, Upton B, Lee SY, Ewing L, Zhang B, Danzer SC and Loepke AW: Effect of dexmedetomidine on sevoflurane-induced neurodegeneration in neonatal rats. Br J Anaesth. 126:1009–1021. 2021. View Article : Google Scholar : PubMed/NCBI

175 

Fu Y and Jin Z: Effects of dexmedetomidine on cognitive function, oxidative stress and brain protection in patients undergoing craniocerebral surgery. Actas Esp Psiquiatr. 52:19–27. 2024. View Article : Google Scholar : PubMed/NCBI

176 

Shan Y, Yang F, Tang Z, Bi C, Sun S, Zhang Y and Liu H: Dexmedetomidine ameliorates the neurotoxicity of sevoflurane on the immature brain through the BMP/SMAD signaling pathway. Front Neurosci. 12:9642018. View Article : Google Scholar

177 

Williams B, Zou L, Pittet JF and Chao W: Sepsis-Induced coagulopathy: A comprehensive narrative review of pathophysiology, clinical presentation, diagnosis, and management strategies. Anesth Analg. 138:696–711. 2024. View Article : Google Scholar : PubMed/NCBI

178 

Chen Z, Zhang H, Qu M, Nan K, Cao H, Cata JP, Chen W and Miao C: Review: The emerging role of neutrophil extracellular traps in sepsis and sepsis-associated thrombosis. Front Cell Infect Microbiol. 11:6532282021. View Article : Google Scholar : PubMed/NCBI

179 

Hagel S, Bach F, Brenner T, Bracht H, Brinkmann A, Annecke T, Hohn A, Weigand M, Michels G, Kluge S, et al: Effect of therapeutic drug monitoring-based dose optimization of piperacillin/tazobactam on sepsis-related organ dysfunction in patients with sepsis: A randomized controlled trial. Intensive Care Med. 48:311–321. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Liu M, Duan S, Zhao X, Wang Q, Yang S, Ma N and Li X: Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review). Int J Mol Med 55: 94, 2025.
APA
Liu, M., Duan, S., Zhao, X., Wang, Q., Yang, S., Ma, N., & Li, X. (2025). Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review). International Journal of Molecular Medicine, 55, 94. https://doi.org/10.3892/ijmm.2025.5535
MLA
Liu, M., Duan, S., Zhao, X., Wang, Q., Yang, S., Ma, N., Li, X."Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review)". International Journal of Molecular Medicine 55.6 (2025): 94.
Chicago
Liu, M., Duan, S., Zhao, X., Wang, Q., Yang, S., Ma, N., Li, X."Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review)". International Journal of Molecular Medicine 55, no. 6 (2025): 94. https://doi.org/10.3892/ijmm.2025.5535
Copy and paste a formatted citation
x
Spandidos Publications style
Liu M, Duan S, Zhao X, Wang Q, Yang S, Ma N and Li X: Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review). Int J Mol Med 55: 94, 2025.
APA
Liu, M., Duan, S., Zhao, X., Wang, Q., Yang, S., Ma, N., & Li, X. (2025). Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review). International Journal of Molecular Medicine, 55, 94. https://doi.org/10.3892/ijmm.2025.5535
MLA
Liu, M., Duan, S., Zhao, X., Wang, Q., Yang, S., Ma, N., Li, X."Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review)". International Journal of Molecular Medicine 55.6 (2025): 94.
Chicago
Liu, M., Duan, S., Zhao, X., Wang, Q., Yang, S., Ma, N., Li, X."Research status and advances in dexmedetomidine for sepsis‑induced multiple organ dysfunction syndrome (Review)". International Journal of Molecular Medicine 55, no. 6 (2025): 94. https://doi.org/10.3892/ijmm.2025.5535
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team