Blog

  • Prevalence and Factors Associated with Rapid Eye Movement-Related Obst

    Prevalence and Factors Associated with Rapid Eye Movement-Related Obst

    Introduction

    Narcolepsy is a chronic neurological disorder with an autoimmune basis, affecting 25–50 per 100,000 individuals worldwide.1 It is primarily characterized by excessive daytime sleepiness (EDS), cataplexy, sleep paralysis, fragmented nighttime sleep, and hypnagogic hallucinations.2 Narcolepsy is classified into two types: Type 1 (NT1), which includes cataplexy (the sudden loss of muscle tone in response to strong emotions), and Type 2 (NT2), which does not.2 EDS, the hallmark symptom of narcolepsy, can also occur in other, more common sleep disorders, such as obstructive sleep apnea (OSA).3 This overlap can complicate the diagnosis and management of narcolepsy.4

    OSA is a prevalent sleep disorder characterized by repeated episodes of upper airway obstruction during sleep, leading to intermittent hypoxia and sleep fragmentation.5,6 Its prevalence ranges from 9% to 38%, with a higher predominance in men.5 While OSA events can occur during any sleep stage, they are typically more severe and prolonged during rapid eye movement (REM) sleep, resulting in significant oxygen desaturation and increased frequency of arousals.7 This REM-specific vulnerability is associated with increased sympathetic stimulation, cardiovascular instability, and diminished vagal tone, potentially leading to adverse cardiovascular events such as hypertension, left ventricular hypertrophy, angina, and myocardial infarction.8–11

    Narcolepsy and OSA may coexist, and both conditions share overlapping features such as EDS and elevated body mass index (BMI). These similarities can delay the diagnosis and complicate the management of narcolepsy, which is less common than OSA.12–15 Therefore, clinicians are advised to assess for the unique symptoms of narcolepsy, particularly cataplexy, in patients presenting with OSA symptoms. Importantly, continuous positive airway pressure (CPAP) therapy, the standard treatment for OSA, does not necessarily resolve EDS in patients with comorbid narcolepsy and OSA.3,4 Thus, proper diagnosis and management of narcolepsy are critical for controlling EDS, the most disabling symptom associated with poor outcomes.

    Although the relationship between narcolepsy and OSA has been studied,4,16 data on the specific association between narcolepsy and REM-related OSA remain limited. For clarity and brevity, we use the term REM-OSA throughout this manuscript to refer to patients who meet our study definition of REM-related OSA. REM-OSA represents a distinct phenotype of sleep-disordered breathing that predominantly affects younger women and involves respiratory events concentrated during REM sleep.10,17

    Recent studies suggest that orexin deficiency in narcolepsy may predispose to REM-related respiratory instability through impaired upper airway regulation.18 Orexin deficiency disrupts muscle tone control during REM sleep, particularly affecting upper airway stability, which may increase the risk of REM-OSA.19,20 Studies have shown altered upper airway muscle activity during REM sleep in orexin-deficient models and in humans with narcolepsy.19,20 Beyond muscle tone effects, fragmented sleep patterns common in narcolepsy create additional respiratory vulnerabilities. Sleep fragmentation disrupts normal ventilatory control mechanisms, while frequent sleep–wake transitions may compromise upper airway compensatory responses.21,22 Moreover, autonomic dysfunction associated with orexin deficiency affects cardiovascular and respiratory regulation, potentially exacerbating sleep-disordered breathing episodes.23 Exaggerated REM muscle atonia and fragmented sleep characteristic of narcolepsy may worsen respiratory events during this sleep stage.24 However, the exact mechanisms linking orexin loss, changes in REM sleep, and airway collapse remain complex and under investigation.10,25 Moreover, the temporal relationship between REM sleep and respiratory events in narcolepsy remains poorly understood, particularly given the altered REM sleep architecture characteristic of this disorder. Clarifying these relationships could guide treatments targeting sleep disordered breathing in narcolepsy.18

    While Hoshino et al reported REM-OSA prevalence in Japanese patients with narcolepsy, their study was descriptive and lacked matched controls and was limited to a single population.26 Our study addresses these gaps by including matched controls and representing the first investigation of REM-OSA in narcolepsy from the Middle East, expanding global understanding across diverse populations.

    This study aims to assess the prevalence and identify factors independently associated with OSA and REM-OSA in patients diagnosed with narcolepsy. In addition, we included a control group of individuals with OSA but without narcolepsy, matched for age, sex, AHI, and BMI, to compare the distribution of REM-OSA between the two groups. Specifically, we hypothesize that those diagnosed with narcolepsy have higher rates of REM-OSA compared to matched controls. Given the known orexin deficiency in NT1, we further hypothesize that NT1 patients may demonstrate different REM-OSA patterns compared to NT2 patients. This investigation seeks to inform evidence-based screening protocols and treatment algorithms for sleep-disordered breathing in narcolepsy populations, ultimately improving patient outcomes through more targeted therapeutic interventions.

    Materials and Methods

    This retrospective analysis was conducted using prospectively collected cohort data from the University Sleep Disorders Center (USDC) research database at King Saud University Medical City, Riyadh, Saudi Arabia.27 The USDC has maintained a comprehensive prospective research database, systematically capturing demographic, clinical, and polysomnographic data using predefined forms and standardized protocols. Trained personnel entered data prospectively at the time of clinical evaluation, with continuous quality control measures in place throughout the study period.28,29

    The current analysis included all consecutive adult patients (≥18 years) diagnosed with narcolepsy between January 2007 and February 2022, identified through systematic database queries using International Classification of Sleep Disorders, 3rd edition (ICSD-3) diagnostic criteria. This approach ensured comprehensive case identification while minimizing selection bias inherent in retrospective analyses.30

    The study received approval from the Institutional Review Board (IRB) at King Saud University Medical City (Approval Number: 19/0134IRB). Informed consent was obtained from all patients for clinical evaluation and data collection at the time of their initial assessment, with additional consent for research use of de-identified data when required by institutional policy.

    Participants and Selection Criteria

    The following criteria were applied to include patients with narcolepsy in the original recruitment protocol for the cohort: (1) age ≥18 years at the time of diagnosis; (2) confirmed diagnosis of NT1 or NT2 according to ICSD-3 criteria; (3) completion of overnight polysomnography and MSLT; and (4) availability of complete demographic and anthropometric data. Exclusion criteria included: (1) incomplete polysomnographic studies due to technical issues or insufficient sleep time (<6 hours total sleep time); (2) concurrent use of medications at the time of the sleep study or within 4 weeks of the study known to significantly affect REM sleep architecture (such as antidepressants or stimulants); and (3) significant medical comorbidities that could affect sleep architecture, including active psychiatric disorders requiring hospitalization or severe cardiopulmonary disease.

    Sample Size Justification

    The sample comprised 190 patients with narcolepsy, representing the entire population of diagnosed cases at our center during the 15-year study period, ensuring comprehensive case identification and minimizing selection bias. This approach is consistent with best practices for retrospective studies utilizing complete case ascertainment from defined clinical populations.31 The inclusion of 122 matched controls with OSA but without narcolepsy provided reasonable comparison data for detecting potentially meaningful differences in REM-OSA distribution between groups.

    Our study design improves upon previous narcolepsy-REM-OSA research by incorporating a matched control group, which was absent in the seminal work by Hoshino et al.26 This methodological advancement enables direct comparative analysis and strengthens the inference of a potential association between narcolepsy and REM-OSA development.

    Recent population-based data from Saudi Arabia show that REM-OSA affects 2.68% of adults overall and 30.64% of patients with OSA.32 These benchmarks informed our power calculations and interpretation. Our tertiary-center narcolepsy cohort comprised 190 consecutive adults; 106 (55.8%) met the diagnostic criteria for OSA and thus entered the REM-OSA analysis set.

    Although formal a priori power calculations were not feasible due to the retrospective design, we conducted a post-hoc power analysis for interpretive context. This analysis indicated approximately 83% power to detect the observed 9.2% difference in REM-OSA prevalence between narcolepsy patients and matched controls (Cohen’s h = 0.22). We emphasize, however, that confidence intervals and effect sizes are more informative than significance testing in interpreting this finding.

    The post-hoc power analysis is presented for interpretive context only. It does not alter the non-significant result, which should be interpreted primarily through confidence intervals and effect sizes.

    Clinical Assessment and Data Collection

    All patients completed a questionnaire assessing narcolepsy-related symptoms, including irresistible attacks of sleep as well as cataplexy, sleep paralysis, and sleep-related hallucinations. Additional information on comorbid conditions, including OSA symptoms, past medical history, and medication use, was also collected. The questionnaire was administered by trained sleep technologists and reviewed by sleep medicine physicians to ensure accuracy and completeness of symptom reporting.

    Demographic and anthropometric measurements, such as age, sex, weight, height, neck circumference, waist and hip circumferences, blood pressure, pulse, and BMI, were recorded at the initial visit. All anthropometric measurements were performed using standardized techniques: height was measured to the nearest 0.1 cm using a wall-mounted stadiometer, weight was recorded to the nearest 0.1 kg using a calibrated digital scale, and neck circumference was measured at the level of the cricothyroid membrane.

    Only data elements explicitly outlined in the approved protocol and relevant to the study’s aims regarding REM-OSA prevalence and associated factors were extracted and made available for analysis.

    Polysomnographic and MSLT Procedures

    In-lab PSG and MSLT were performed based on clinical assessment. All polysomnographic studies were conducted using standardized equipment (Alice-4 and Alice-6 systems, Philips Respironics, Murrysville, PA, USA) and scored according to American Academy of Sleep Medicine (AASM) criteria.33 Sleep stages were scored in 30-second epochs, and respiratory events were identified using standard AASM definitions. Hypopneas were defined as ≥30% reduction in airflow lasting ≥10 seconds associated with either ≥3% oxygen desaturation or arousal. Apneas were defined as ≥90% reduction in airflow for ≥10 seconds. The polysomnographic parameters analyzed included the apnea-hypopnea index (AHI), arousal index, mean oxygen saturation, minimum oxygen saturation, and sleep-onset REM periods (SOREMP). MSLT was performed the day following polysomnography according to the standard protocol, with five 20-minute nap opportunities at 2-hour intervals beginning 1.5–3 hours after morning awakening.34

    Diagnostic Criteria

    Narcolepsy and OSA were diagnosed according to the criteria outlined in the International Classification of Sleep Disorders, 3rd edition.2 All patients met the requirement of having daily periods of irrepressible need to sleep or daytime sleep lapses for at least three months. NT1 was diagnosed if cataplexy was present along with a mean sleep latency ≤8 minutes and ≥2 sleep-onset REM periods (SOREMPs) on a standard MSLT, with a SOREMP on the preceding PSG allowed to substitute for one MSLT SOREMP. Alternatively, NT1 was also diagnosed if cerebrospinal fluid (CSF) hypocretin-1 concentration was ≤110 pg/mL or <1/3 of the mean normal values. NT2 was diagnosed when cataplexy was absent, the MSLT showed a mean sleep latency of ≤8 minutes and ≥2 SOREMPs, CSF hypocretin-1 was either not measured or >110 pg/mL (or >1/3 of the normal mean), and the findings could not be better explained by other causes of hypersomnolence. OSA was defined as AHI ≥5 events/hour of sleep accompanied by symptoms or comorbidities, or AHI ≥15 events/hour regardless of symptoms.

    OSA was further categorized into REM-OSA and non-stage-specific (NSS-OSA). REM-OSA was defined by an AHI ≥5, an AHI-REM/AHI-NREM ratio ≥2, an AHI-NREM <8, and REM sleep duration exceeding 10.5 minutes.26,35,36

     This definition was selected to ensure specificity and avoid misclassification of patients with minimal REM sleep, as shorter REM periods may not provide adequate time for reliable assessment of REM-specific respiratory events.10

    Control Group Selection

    To allow comparative analysis, a control group of 122 adult patients diagnosed with OSA but without narcolepsy was included. Controls were selected from the same sleep disorders center database and matched to the narcolepsy group based on age, sex, AHI, and BMI. Matching was performed using a 1:1 nearest neighbor approach with a caliper width of 0.2 standard deviations for continuous variables. When multiple potential matches were available, controls were selected randomly to minimize selection bias. All control patients underwent overnight PSG during the same study period and met the diagnostic criteria for OSA according to AASM guidelines. Narcolepsy was excluded in the control group based on clinical assessment and the absence of MSLT. Standardized mean differences for age, sex, BMI, and AHI were all below 0.1 after matching, indicating successful balance between cases and controls. Additional exclusion criteria for controls included: (1) reported cataplexy-like episodes; and (2) documented sleep-onset REM periods during routine polysomnography. The control group was used to compare the prevalence and distribution of REM-OSA between patients with and without narcolepsy.

    Control Group Rationale

    We chose matched patients with OSA but without narcolepsy as the comparison group, rather than healthy sleepers, for three reasons. First, using disease controls allows us to isolate the incremental contribution of narcolepsy-related pathophysiology to REM-OSA while holding constant the baseline impact of upper-airway obstruction. Second, OSA is highly prevalent in narcolepsy; therefore, comparing narcolepsy + OSA with “OSA-only” patients addresses a question that clinicians face daily, namely, whether narcolepsy alters the OSA phenotype. Third, frequency-matching on AHI removes a major confounder; without this step, differing OSA severities would obscure any narcolepsy-specific effects.

    While this design cannot establish absolute REM-OSA prevalence in narcolepsy, it provides a good, rigorous approach to isolate narcolepsy-specific contributions to REM-OSA.

    Statistical Analysis

    Descriptive statistics summarized demographic and clinical characteristics. Continuous variables are reported as mean ± SD and compared with independent t-tests (Welch t-test when variances were unequal); when Shapiro–Wilk or histogram inspection indicated non-normality, the Mann–Whitney U-test was applied. Normality assumptions for t-tested variables were verified statistically (Shapiro–Wilk test). Categorical variables were presented as frequencies (%) and compared with Pearson’s χ²-test; when any expected cell was < 5, Fisher’s exact test was used. Associations between two binary variables (eg, narcolepsy subtype vs overall OSA status) were assessed with Pearson’s χ²-test; the φ coefficient was reported as the effect size. For the matched comparison of REM-OSA vs NSS-OSA, group differences were evaluated with conditional logistic regression (strata = age–sex–BMI triplets), and McNemar’s exact test was used for the paired 2×2 counts. Multivariable logistic regression identified factors independently associated with OSA and REM-OSA; candidate variables entered the model if univariable p < 0.10 or strong clinical relevance was present. Models were restricted to ≥ 8 outcome events per variable (EPV) to limit over-fitting, following current methodological recommendations.37,38 Model fit was assessed with the Hosmer–Lemeshow goodness-of-fit test, discrimination with the area under the ROC curve, explained variance with Nagelkerke R², and multicollinearity with variance-inflation factors (VIF < 5). Effect sizes were expressed for the primary comparisons using Cohen’s conventions—d for continuous variables, h for proportions, and φ for binary associations. All descriptive and comparative analyses were executed in SPSS v25.0 (IBM Corp., Armonk, NY, USA). A post-hoc power analysis was performed for interpretive context only, showing ~83% power to detect the observed small-to-medium effect size (Cohen’s h = 0.22). This analysis does not affect the interpretation of non-significant findings.39,40 This yielded 83% power to detect a two-sided difference at α = 0.05. Binomial 95% confidence intervals for single proportions were obtained with the Wilson score method.41,42

    The original cohort was assembled prospectively, and every study variable was captured for each participant; the analytic dataset was therefore complete (with no missing data), and no imputation was required.

    All statistical tests were considered exploratory; no correction for multiple comparisons was applied, and findings should be interpreted accordingly.

    Results

    This study investigated the prevalence and identified factors independently associated with OSA and its phenotype, REM-OSA, among patients with NT1 and NT2, and compared the distribution of REM-OSA with a matched control group of patients with OSA but without narcolepsy. A total of 190 patients were diagnosed with narcolepsy, including 152 males (80%) and 38 females (20%). Among them, 119 patients (62.6%) were diagnosed with NT1, and 71 patients (37.4%) were diagnosed with NT2. The classification and distribution of OSA and its phenotypes among NT1 and NT2 are illustrated in Figure 1. Narcolepsy subtypes demonstrated remarkable similarity in both demographic characteristics and sleep-disordered breathing patterns. The sex distribution showed no significant difference between the NT1 and NT2 groups (OR: 0.62, 95% CI: 0.29–1.35, p = 0.31), although our tertiary care setting’s male predominance may reflect referral patterns rather than the true population prevalence (Table 1).

    Table 1 Prevalence and Distribution of OSA, REM-OSA, and NSS-OSA Among Patients with Narcolepsy (NT1 Vs NT2)

    Figure 1 Flowchart of OSA, REM-Related OSA, and NSS-OSA Classification Among Patients with Narcolepsy.

    Abbreviations: OSA, obstructive sleep apnea; REM-related OSA, rapid eye movement–related obstructive sleep apnea; NSS-OSA, non-stage-specific obstructive sleep apnea.

    Most importantly, OSA prevalence was virtually identical between narcolepsy subtypes (55.5% vs 56.3%, OR: 0.97, 95% CI: 0.54–1.74, p = 1.00), suggesting that OSA risk in narcolepsy operates independently of cataplexy presence and orexin deficiency status. Among the 106 narcolepsy patients who had OSA, 28 fulfilled REM-OSA criteria (26.4%; 95% CI 18.9–35.5%, Wilson) (Figure 2). REM-OSA showed a non-significant trend toward higher prevalence in NT2 (30.0% vs 24.2%, OR: 0.75, 95% CI: 0.31–1.80, p = 0.67), though the wide confidence interval reflects limited statistical power for this subgroup analysis (Table 1).

    Figure 2 Prevalence of REM-OSA. Bars show the proportion of patients with REM-OSA among the pooled narcolepsy cohort (green), controls (blue), narcolepsy type 1 subtype (purple) and narcolepsy type 2 subtype (Orange). The double-headed arrow depicts the standardized difference between the pooled narcolepsy cohort and controls (Cohen’s h = 0.22, small–to-medium effect).

    Table 2 summarizes the demographic and polysomnographic characteristics of patients with narcolepsy with versus without OSA. Patients with OSA had a higher BMI (31.5 ± 7.2 vs 28.3 ± 5.9 kg m², p = 0.001, Mann–Whitney U), and an elevated arousal index (29.3 ± 20.1 vs 13.8 ± 7.7 events/hour, p < 0.001). As expected, respiratory event indices and oxygen-desaturation metrics were also markedly higher in the OSA subgroup during both REM and NREM sleep (all p < 0.001). To interpret the clinical importance of observed group differences, effect sizes were calculated. Effect-size estimates revealed moderate to very large differences between patients with narcolepsy with and without OSA, including a moderate difference in BMI (Cohen’s d = 0.48) and a large difference in arousal index (d = 1.02). These findings underscore the clinical relevance of variables such as BMI, arousal index, and respiratory-event indices (Table 2).

    Table 2 Clinical and Polysomnographic Characteristics of Patients with Narcolepsy with and Without Obstructive Sleep Apnea (N = 190)

    Age and AHI were approximately normally distributed (Shapiro–Wilk p > 0.10) and were compared with independent t-tests, whereas BMI showed mild right skew and was analyzed with the Mann–Whitney U-test. No significant differences emerged between NT1 and NT2 (Age: 31.2 ± 10.8 vs 32.0 ± 11.1 years, p = 0.55; BMI: 28.9 ± 4.6 vs 29.3 ± 4.2 kg m², p = 0.62; AHI: 23.8 ± 14.7 vs 24.4 ± 15.1 events/hours, p = 0.80). Cohen’s d values for age (0.07) and AHI (0.05) were also ≤ 0.10, confirming that the observed differences are clinically negligible.

    Regarding narcolepsy symptoms, the OSA group reported higher frequencies of hypnagogic and hypnopompic hallucinations, fragmented sleep, and sleep paralysis; however, these differences were not statistically significant. Comorbid conditions such as hypertension, diabetes mellitus, and bronchial asthma were more prevalent among patients with narcolepsy and OSA compared to those without, though these differences did not reach statistical significance (Table 2).

    Logistic Regression Analysis

    Logistic regression analysis revealed several factors independently associated with OSA and its phenotype, REM-OSA in narcolepsy (Table 3). For overall OSA, male sex conferred a more than fourfold increase in risk (OR = 4.45, 95% CI 1.44–13.73, p < 0.01), while each additional kilogram per square-meter of BMI raised the odds by 7% (OR = 1.07, 95% CI 1.00–1.15, p = 0.05). A higher arousal index was likewise associated with greater OSA probability (OR = 1.12, 95% CI 1.07–1.17, p < 0.01). Within the OSA subgroup, REM-OSA was independently associated with the arousal index (OR = 1.13, 95% CI 1.06–1.19, p < 0.01), whereas longer REM-sleep duration exerted a modest protective effect (OR = 0.98, 95% CI 0.97–0.99, p < 0.01). Pearson’s χ²-tests found no association between narcolepsy subtype (NT1 vs NT2) and either overall OSA status (χ² = 0.66, df = 1, p = 0.42; φ = 0.06) or the distribution of REM-OSA versus non-stage-specific OSA (χ² = 0.01, df = 1, p = 0.91; φ = 0.01). Model diagnostics supported the robustness of these findings: the OSA model showed good calibration (Hosmer–Lemeshow χ² = 7.2, df = 8, p = 0.51) and strong discrimination (AUC = 0.84, 95% CI 0.78–0.90), while the REM-OSA model also fit adequately (Hosmer–Lemeshow χ² = 5.4, df = 8, p = 0.71) with moderate discrimination (AUC = 0.73, 95% CI 0.63–0.82). Variance-inflation factors for all correlates were below 1.8, indicating that multicollinearity was not a concern.

    Table 3 Factors Associated with OSA and REM-OSA Among Patients with Narcolepsy (Logistic Regression Analysis)

    Comparison with Control Group

    To characterize REM-OSA in narcolepsy, we compared 106 narcolepsy patients with OSA to 122 age-, sex-, AHI-, and BMI-matched OSA controls without narcolepsy (Tables 4 and 5). Among controls, 21/122 had REM-OSA (17.2%; 95% CI 11.0–25.1%), whereas in the narcolepsy group 28/106 had REM-OSA (26.4%; 95% CI 18.9–35.5%). The unadjusted odds ratio (OR) was 1.73 (95% CI 0.91–3.27, p = 0.09; see Table 4), with Cohen’s h = 0.22 (small–medium effect). A conditional logistic model that preserved the matching yielded a similar estimate (OR = 1.69, 95% CI 0.89–3.18, p = 0.11); for comparison with earlier unmatched studies, the unmatched Pearson χ² (2.85, p = 0.09) is also shown in Table 4. McNemar’s exact test was concordant (p = 0.12). Because none of these tests crossed the 0.05 threshold, the higher REM-OSA prevalence in narcolepsy should be viewed as a non-significant trend; with only 28 events, the study may be underpowered to detect a modest effect.

    Table 4 Comparison of REM-OSA and Non-Stage Specific OSA Between Patients with Narcolepsy and the Control Group (Unadjusted Comparison)

    Table 5 Sex Distribution and REM-OSA Phenotype Characteristics in Cases with Narcolepsy and Controls

    Sex distribution within the REM-OSA subgroup showed a similar male predominance in both groups, with males representing 76.2% in the control group and 82.1% in the narcolepsy group (Table 5).

    Discussion

    This is the first controlled investigation of REM-OSA in patients with narcolepsy, representing a significant methodological advancement over previous descriptive studies through the inclusion of age-, sex-, AHI-, and BMI-matched controls. In the present study, OSA was found to be prevalent in both types of narcolepsy, with more than half of the patients (55.8%) diagnosed with OSA. These results are consistent with previous reports that suggest an increased prevalence of OSA among patients with narcolepsy.4,43–45 The coexistence of narcolepsy and OSA may further worsen fragmented sleep and negatively impact the quality of life for these patients.46,47

    Several factors may explain the high prevalence of OSA observed in our study. Notably, BMI, a well-established risk factor for OSA, was elevated in our cohort. The average BMI of 31.5 ± 7.2 classifies the majority of patients as overweight or obese, which likely contributed to the increased occurrence of OSA. Other factors may also elevate OSA risk in patients with narcolepsy. For example, low orexin levels in patients with NT1 could attenuate the ventilatory response to hypercapnia and intermittent hypoxia.18,48 A poor response to these conditions may result in inadequate activation of the muscles that maintain upper airway patency, increasing the risk of airway collapse during sleep. Experimental studies have shown that orexin A and B enhance the activity of the genioglossus muscle, a major upper airway dilator muscle.49,50 Consequently, orexin deficiency may impair genioglossus muscle function, especially in REM sleep when muscle tone is naturally reduced, thereby increasing the risk of REM-OSA.10

    The comparative analysis between NT1 and NT2 revealed several unexpected findings that challenge the conventional understanding of narcolepsy pathophysiology. The virtually identical OSA prevalence between subtypes (55.5% vs 56.3%, p = 1.00) suggests that sleep-disordered breathing risk in narcolepsy may be independent of orexin deficiency status. This finding contradicts the previously discussed hypothesis linking orexin deficiency to greater respiratory instability during sleep.49

    There is a lack of unified criteria for diagnosing REM-OSA, and significant heterogeneity exists in its definition; therefore, varying prevalence rates, natural history, and clinical significance have been reported in the literature.10 We used a strict definition of REM-OSA that includes REM duration (defined as an AHI ≥5, AHI-REM/AHI-NREM ≥2, AHI-NREM <8, and REM sleep duration >10.5 minutes) to avoid misclassification bias and ensure that our findings accurately represent the true prevalence of REM-OSA in our sample. Despite using this precise definition, REM-OSA was observed in 26.4% of patients with narcolepsy and OSA. Recent population-based studies have reported REM-OSA prevalence ranging from 2.7% in the general population to 30.6% among patients with OSA, providing important context for interpreting our findings.32 The 26.4% REM-OSA prevalence in our narcolepsy cohort falls within the upper range of these estimates, supporting the hypothesis that narcolepsy may predispose to REM-related respiratory events.

    Moreover, our findings align with and significantly extend the work of Hoshino et al,26 who reported REM-OSA prevalence of 25.6–47.1% in Japanese patients with narcolepsy, depending on the diagnostic criteria used. However, our study provides several methodological advances that strengthen the evidence base: (1) inclusion of matched controls enabling direct comparative analysis, (2) larger sample size with 190 patients with narcolepsy versus 141 in the Japanese study, (3) representation of a previously unstudied Middle Eastern population, and (4) rigorous statistical methodology including conditional logistic regression and comprehensive effect size reporting.

    REM-OSA prevalence did not differ significantly between narcolepsy subtypes (30.0% in NT2 vs 24.2% in NT1, χ² = 0.01, p = 0.67), a finding that challenges the expectation of greater REM-related respiratory instability in orexin-deficient NT1. If orexin deficiency were the primary driver of REM-related respiratory events, one would expect higher REM-OSA rates in NT1 patients. However, our findings suggest alternative mechanisms may be responsible for REM-OSA development in narcolepsy. Recent research has identified multiple pathways through which REM sleep and narcolepsy may influence respiratory control, including altered sleep architecture and modified arousal thresholds, which predispose to respiratory instability.10,51 These mechanisms may operate independently of orexin status, potentially explaining the similar OSA risk across narcolepsy subtypes. Clinicians should maintain equal vigilance for OSA symptoms in both NT1 and NT2, as the presence or absence of cataplexy does not appear to influence the burden of respiratory comorbidity. Given that EDS is universal in narcolepsy, it is important to consider that coexisting OSA may contribute to or worsen this symptom, underscoring the need for polysomnographic evaluation regardless of narcolepsy subtype.

    Beyond orexin pathways, emerging research suggests that sleep fragmentation itself may play a role in REM-OSA development in narcolepsy. The characteristic sleep instability in narcolepsy, evidenced by frequent sleep-onset REM periods and disrupted sleep architecture, may create conditions that favor REM-specific respiratory events.52 Additionally, the altered arousal threshold in patients with REM-OSA might influence the termination of respiratory events, potentially prolonging apneas during REM sleep when arousal mechanisms are naturally suppressed.53 These findings suggest that therapeutic interventions targeting sleep consolidation, in addition to traditional OSA treatments, may be particularly beneficial in this population.

    While REM-OSA is often reported to be prevalent among middle-aged female patients, the majority of our study population was male (80%), yet REM-OSA remained prevalent. This demographic pattern may reflect referral bias in our tertiary care setting, where males with narcolepsy symptoms might be more likely to seek medical attention or be referred for sleep evaluation. However, the persistent high REM-OSA prevalence despite male predominance suggests that narcolepsy-related mechanisms may override typical gender-based risk patterns observed in general OSA populations, strengthening the hypothesis for an intrinsic pathophysiological link.

    The geographic and ethnic diversity represented by our Middle Eastern cohort addresses a critical gap in narcolepsy research, which has been predominantly conducted in Asian, European, and North American populations. Given the potential influence of genetic, environmental, and lifestyle factors on sleep-disordered breathing patterns, our findings provide essential validation of REM-OSA prevalence in narcolepsy across different populations.54

    Our study found that male sex, higher BMI, and elevated arousal index were significantly associated with OSA, consistent with established risk factors such as male predominance and obesity. Arousal index, an indicator of sleep fragmentation, also emerged as a significant correlate of REM-OSA. Elevated arousal index reflects frequent sleep interruptions and non-restorative sleep, which may contribute to EDS, even when total sleep time appears normal, and may exacerbate irresistible sleep attacks in individuals with narcolepsy.55 Moreover, previous studies have identified the arousal index as a predictor of carotid atherosclerosis in patients with OSA,56 underscoring the importance of treating OSA effectively. In our study, longer REM sleep duration was associated with a decreased risk of REM-OSA, possibly suggesting that patients with more stable REM sleep are less likely to experience REM-related apneas.

    Comparison with a control group of patients with OSA but without narcolepsy revealed a trend toward higher REM-OSA prevalence among those with narcolepsy (26.4% vs 17.2%, OR: 1.73, 95% CI: 0.91–3.27, p = 0.09). While this difference did not achieve conventional statistical significance, the effect size (Cohen’s h = 0.22) suggests a small to medium effect that may have clinical relevance. This trend did not meet conventional significance thresholds and should be viewed as hypothesis-generating, warranting validation in larger studies. The wide confidence interval reflects the inherent challenges of studying rare sleep disorders. It indicates that larger, multicenter studies are needed to definitively establish whether narcolepsy contributes to REM-OSA development independent of traditional risk factors such as age, sex, and BMI. The observed trend aligns with theoretical considerations regarding altered REM sleep architecture in narcolepsy, though the current sample size may be insufficient to detect this association with adequate statistical power.10

    Narcolepsy, OSA, and its subtype REM-OSA have all been associated with increased cardiometabolic risks.57–59 The simultaneous presence of these conditions may have a synergetic effect, leading to worse cardiometabolic outcomes. This mandates close monitoring of cardiometabolic comorbidities and the concurrent treatment of both narcolepsy and OSA to optimize patient outcomes.

    Our identification of independent associations between narcolepsy and REM-OSA supports the notion that sleep-disordered breathing subtypes warrant distinct clinical consideration, as REM-OSA has been found to be associated with increased risk of hypertension and metabolic complications such as insulin resistance and metabolic syndrome.10 Recognizing REM-OSA in patients with narcolepsy warrants tailored evaluation and management to mitigate sleep fragmentation and improve daytime functioning through targeted interventions. These may include optimizing positive airway pressure therapy to address REM-specific obstruction and exploring adjunctive pharmacologic strategies that stabilize REM sleep or enhance upper airway muscle tone.60 The clinical implications of our findings therefore extend beyond diagnostic considerations to treatment strategies. Current evidence suggests that patients with narcolepsy may respond differently to CPAP therapy compared to those with OSA alone, potentially requiring modified treatment approaches or adjunctive therapies.4 Furthermore, the presence of REM-OSA in patients with narcolepsy may necessitate careful titration of CPAP pressures, as REM-specific respiratory events often require higher therapeutic pressures than non-REM events.10 The interaction between narcolepsy medications and OSA treatment also warrants consideration, as stimulants commonly used for EDS may affect sleep architecture and potentially influence OSA severity.61 Current evidence suggests that adherence to positive airway pressure (PAP) therapy in patients with REM-OSA is suboptimal, and the currently accepted criteria for good adherence to PAP therapy, 4 hours per night, may not be suitable for REM-OSA, as they do not cover most of the REM sleep periods.10 Clinical management should consider potential confounding effects of narcolepsy treatments, such as sodium oxybate, which may exacerbate sleep-disordered breathing, highlighting the need for close monitoring and multidisciplinary care, though treatment with CPAP has been shown to normalize AHI in patients while continuing therapy with sodium oxybate.62 Although prospective trials are needed, emerging evidence suggests that combined therapeutic approaches may yield superior outcomes through phenotypic clustering approaches that leverage the heterogeneity of OSA by classifying it into smaller, more homogeneous disorder subtypes, with approximately half of patients with narcolepsy with comorbid OSA who are adherent to PAP therapy showing improvements in EDS and sleep quality, though responses vary widely.62,63 These complexities underscore the importance of integrating polysomnographic phenotyping with personalized patient care in a multidisciplinary setting, as OSA is a complex and heterogeneous disorder where severity criteria based on AHI alone do not capture the diverse spectrum of the condition.60 Our findings thus highlight the clinical promise of REM-OSA phenotyping as a tool to refine management and alleviate disease burden in patients with narcolepsy.

    Future research should prioritize several key areas to advance our understanding of the narcolepsy-REM-OSA relationship. First, longitudinal studies are needed to determine whether REM-OSA development precedes or follows narcolepsy onset, which could inform screening protocols and early intervention strategies. Second, investigations examining the response to different OSA treatments in patients with narcolepsy could guide evidence-based therapeutic approaches. Third, genetic studies exploring shared susceptibility factors between narcolepsy and REM-OSA may reveal novel pathophysiological pathways. Finally, research examining the impact of narcolepsy-specific treatments on OSA severity and REM-OSA prevalence could inform integrated management approaches.

    Several limitations warrant careful consideration. First, the subtype comparison between NT1 and NT2 was strictly exploratory; the present study was not powered to detect smaller inter-subtype differences, and larger multicenter cohorts will be required to definitively address this question. The limited number of REM-OSA events (n = 28) reduced statistical power for subgroup analyses and fell below recommended thresholds for stable logistic regression, potentially inflating variance around these estimates. Nevertheless, the confidence interval around the primary estimate (OR = 1.73, 95% CI: 0.91–3.27) excludes a trivial effect, supporting our interpretation of a small-to-medium association that merits replication in larger samples. Second, the observational, retrospective design inherently limits causal inference and introduces selection bias, a common issue in tertiary sleep clinic populations.31 Third, incomplete orexin measurements restricted exploration of potential mechanisms linking orexin deficiency to REM-OSA, especially given the theoretical role of orexin in upper airway tone regulation during REM sleep. Forth, our single-ethnicity cohort from the Middle East limits generalizability to populations with different genetic backgrounds and environmental exposures. Fifth, sampling controls from the same tertiary center introduces referral bias, as both groups reflect specialized care-seeking populations rather than community samples.28 Residual confounding remains possible because variables like detailed sleep architecture metrics and subclinical narcolepsy features were not matched. Additionally, heterogeneity in REM-OSA definitions complicates direct comparisons despite our stringent criteria. Furthermore, detailed REM sleep parameters (such as REM density, REM latency, and total REM duration) and specific sleep fragmentation indices (including apnea arousal index and PLMI arousal index) were not systematically extracted as part of the originally approved protocol. Future prospective studies incorporating these comprehensive sleep architecture parameters would be valuable for elucidating the detailed mechanistic pathways linking narcolepsy to REM-OSA. Finally, we did not assess treatment response or long-term outcomes, limiting direct clinical applicability.

    Our choice of OSA patients as controls rather than healthy sleepers enhances internal validity for characterizing REM-OSA phenotype in narcolepsy by addressing clinically relevant dual pathology and controlling for the baseline effects of sleep-disordered breathing. However, this design limits conclusions about absolute REM-OSA prevalence in narcolepsy. Future studies with healthy controls and population-based samples are needed to establish baseline REM-OSA rates, particularly given recent evidence of variability in sleep-disordered breathing patterns across clinical populations. Thus, while our findings suggest a potential link between narcolepsy and REM-OSA, they should be viewed as hypothesis-generating and require validation in larger, multicenter studies with adequate power for detecting smaller effect sizes.

    The borderline non-significant REM-OSA difference (χ²=2.85, p=0.09; Cohen’s h=0.22) had ~83% post-hoc power, suggesting a potentially meaningful effect (9.2% prevalence difference) warranting validation in larger studies. Subgroup analyses (NT1 vs NT2) were underpowered (only 28 REM-OSA events, ~9 EPV), falling below the recommended ≥10 EPV for stable logistic regression.37,38 Thus, similar OSA prevalence between NT1 and NT2 should be interpreted cautiously. Multicenter collaborations recruiting larger, diverse cohorts with standardized REM-OSA definitions and comprehensive orexin measures are needed to clarify mechanisms and predictors of sleep-disordered breathing in narcolepsy across different ethnic and geographic populations. We also acknowledge that alternative definitions of REM-related OSA exist and were not tested in sensitivity analyses, which could influence prevalence estimates. Furthermore, given the limited number of REM-related OSA events (~28), small-sample bias remains possible. Future analyses may benefit from penalized regression methods such as Firth correction to address this.

    Conclusion

    This study represents the first controlled investigation of REM-OSA in patients with narcolepsy, advancing beyond previous descriptive reports through matched control design and comprehensive statistical analysis. Using stringent diagnostic criteria, we found a high prevalence of OSA (55.8%) and REM-OSA (26.4%) among patients with narcolepsy. A trend toward higher REM-OSA prevalence was observed compared to matched controls (26.4% vs 17.2%, p = 0.09), suggesting a potential association requiring validation in larger studies.

    Importantly, OSA prevalence was virtually identical between NT1 and NT2 (55.5% vs 56.3%, p = 1.00), suggesting that sleep-disordered breathing risk may operate through mechanisms other than orexin deficiency status. These results emphasize the importance of comprehensive evaluation and tailored management strategies in patients with narcolepsy, while highlighting the need for adequately powered multicenter investigations to definitively establish the narcolepsy-REM-OSA relationship. Finally, observed differences that did not achieve statistical significance should be interpreted as hypothesis-generating and require validation in larger, multicenter cohorts.

    Data Sharing Statement

    Data can be obtained upon request from the corresponding author “ASB”, but this requires institutional approval by the Institutional Review Board of the College of Medicine at King Saud University.

    Ethics Approval and Informed Consent

    The study received approval from the Institutional Review Board (IRB) at King Saud University Medical City (Approval Number: 19/0134IRB). Informed consent was obtained from all patients for clinical evaluation and data collection at the time of their initial assessment, with additional consent for research use of de-identified data when required by institutional policy. For this retrospective analysis, the requirement for additional informed consent was waived by the ethics committee because the study involved anonymized data and posed minimal risk to participants. All procedures involving human participants were conducted in accordance with the ethical standards of the 1964 Declaration of Helsinki and its subsequent revisions.

    Author Contributions

    Hamza O. Dhafar: Conceptualization, Methodology, Writing – original draft preparation, Writing – review and editing, Project administration. Ali A. Awadh: Conceptualization, Writing – original draft preparation, Writing – review and editing. Salih A. Aleissi: Conceptualization, Writing – original draft preparation, Writing – review and editing. Galal Eldin Abbas Eltayeb: Formal analysis, Data curation, Writing – review and editing. Samar Nashwan: Formal analysis, Data curation, Writing – review and editing. Ahmed S. BaHammam: Conceptualization, Writing – original draft preparation, Writing – review and editing, Project administration, Supervision.

    All authors gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agreed to be accountable for all aspects of the work.

    Funding

    The work was funded by the Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia (08-MED511-02).

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Longstreth WT, Koepsell TD, Ton TG, Hendrickson AF, van Belle G. The epidemiology of narcolepsy. Sleep. 2007;30(1):13–26. doi:10.1093/sleep/30.1.13

    2. American Academy of Sleep M. The International Classification of Sleep Disorders: (ICSD-3); 2014.

    3. Bonsignore MR, Pepin JL, Cibella F, et al. Excessive daytime sleepiness in obstructive sleep apnea patients treated with continuous positive airway pressure: data from the European sleep apnea database. Front Neurol. 2021;12.

    4. Sansa G, Iranzo A, Santamaria J. Obstructive sleep apnea in narcolepsy. Sleep Med. 2010;11(1):93–95. doi:10.1016/j.sleep.2009.02.009

    5. Senaratna CV, Perret JL, Lodge CJ, et al. Prevalence of obstructive sleep apnea in the general population: a systematic review. Sleep Med Rev. 2017;34:70–81. doi:10.1016/j.smrv.2016.07.002

    6. Lévy P, Kohler M, McNicholas WT, et al. Correction: obstructive sleep apnoea syndrome. Nat Rev. 2015;1(1). doi:10.1038/nrdp.2015.15

    7. Almeneessier AS, Alshahrani M, Aleissi S, Hammad OS, Olaish AH, BaHammam AS. Comparison between blood pressure during obstructive respiratory events in REM and NREM sleep using pulse transit time. Sci Rep. 2020;10(1):3342. doi:10.1038/s41598-020-60281-2

    8. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural mechanisms in obstructive sleep apnea. J Clin Invest. 1995;96(4):1897–1904. doi:10.1172/JCI118235

    9. Lanfranchi PA, Somers VK. Cardiovascular physiology. Principles Pract Sleep Med. 2011;226–236.

    10. Pirzada AR, BaHammam AS. Rapid eye movement predominant obstructive sleep apnoea: prognostic relevance and clinical approach. Curr Opin Pulm Med. 2021;27(6):514–522. doi:10.1097/MCP.0000000000000817

    11. BaHammam AS, Alshahrani M, Aleissi SA, Olaish AH, Alhassoon MH, Shukr A. Blood pressure dipping during REM and non-REM sleep in patients with moderate to severe obstructive sleep apnea. Sci Rep. 2021;11(1):7990. doi:10.1038/s41598-021-87200-3

    12. Young T, Palta M, Dempsey J, Skatrud J, Weber S, Badr S. The occurrence of sleep-disordered breathing among middle-aged adults. N Engl J Med. 1993;328(17):1230–1235. doi:10.1056/NEJM199304293281704

    13. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol. 2013;177(9):1006–1014. doi:10.1093/aje/kws342

    14. Seneviratne U, Puvanendran K. Excessive daytime sleepiness in obstructive sleep apnea: prevalence, severity, and predictors. Sleep Med. 2004;5(4):339–343. doi:10.1016/j.sleep.2004.01.021

    15. Dhafar HO, BaHammam AS. Body weight and metabolic rate changes in narcolepsy: current knowledge and future directions. Metabolites. 2022;12(11):1120. doi:10.3390/metabo12111120

    16. Pataka AD, Frangulyan RR, Mackay TW, Douglas NJ, Riha RL. Narcolepsy and sleep-disordered breathing. Eur J Neurol. 2012;19(5):696–702. doi:10.1111/j.1468-1331.2011.03610.x

    17. Koo BB, Patel SR, Strohl K, Hoffstein V. Rapid eye movement-related sleep-disordered breathing: influence of age and gender. Chest. 2008;134(6):1156–1161. doi:10.1378/chest.08-1311

    18. Wang Y, Deng T, Zhao X, et al. Control of breathing by orexinergic signaling in the nucleus tractus solitarii. Sci Rep. 2024;14(1):7473. doi:10.1038/s41598-024-58075-x

    19. Tarasiuk A, Levi A, Berdugo-Boura N, Yahalom A, Segev Y. Role of orexin in respiratory and sleep homeostasis during upper airway obstruction in rats. Sleep. 2014;37(5):987–998. doi:10.5665/sleep.3676

    20. Gestreau C, Bévengut M, Dutschmann M. The dual role of the orexin/hypocretin system in modulating wakefulness and respiratory drive. Curr Opin Pulm Med. 2008;14(6):512–518. doi:10.1097/MCP.0b013e32831311d3

    21. Baumann CR, Mignot E, Lammers GJ, et al. Challenges in diagnosing narcolepsy without cataplexy: a consensus statement. Sleep. 2014;37(6):1035–1042. doi:10.5665/sleep.3756

    22. Kubin L. Neural control of the upper airway: respiratory and state-dependent mechanisms. Compr Physiol. 2016;6(4):1801–1850. doi:10.1002/j.2040-4603.2016.tb00729.x

    23. Grimaldi D, Calandra-Buonaura G, Provini F, et al. Abnormal sleep-cardiovascular system interaction in narcolepsy with cataplexy: effects of hypocretin deficiency in humans. Sleep. 2012;35(4):519–528. doi:10.5665/sleep.1738

    24. Miano S, Kheirandish-Gozal L, De Pieri M. Comorbidity of obstructive sleep apnea and narcolepsy: a challenging diagnosis and complex management. Sleep Med. 2024;8:100126.

    25. Spinieli RL, Ben Musa R, Cornelius-Green J, Hasser EM, Cummings KJ. Orexin facilitates the ventilatory and behavioral responses of rats to hypoxia. Am J Physiol Regul Integr Comp Physiol. 2022;322(6):R581–r596. doi:10.1152/ajpregu.00334.2021

    26. Hoshino T, Sasanabe R, Mano M, et al. Prevalence of rapid eye movement-related obstructive sleep apnea in adult narcolepsy. Intern Med. 2019;58(15):2151–2157. doi:10.2169/internalmedicine.2601-18

    27. Almeneessier AS, Alballa NS, Alsalman BH, Aleissi S, Olaish AH, BaHammam AS. A 10-year longitudinal observational study of cataplexy in a cohort of narcolepsy type 1 patients. Nat Sci Sleep. 2019;11:231–239. doi:10.2147/NSS.S229105

    28. Benchimol EI, Smeeth L, Guttmann A, et al. The reporting of studies conducted using observational routinely-collected health data (RECORD) statement. PLoS Med. 2015;12(10):e1001885. doi:10.1371/journal.pmed.1001885

    29. Sedgwick P. Retrospective cohort studies: advantages and disadvantages. BMJ. 2014;348(g1072).

    30. Vandenbroucke JP, von Elm E, Altman DG, et al. Strengthening the reporting of observational studies in epidemiology (STROBE): explanation and elaboration. Int J Surg. 2014;12(12):1500–1524. doi:10.1016/j.ijsu.2014.07.014

    31. Vandenbroucke JP, von Elm E, Altman DG, et al. Strengthening the reporting of observational studies in epidemiology (STROBE): explanation and elaboration. Ann Intern Med. 2007;147(8):W163–194. doi:10.7326/0003-4819-147-8-200710160-00010-w1

    32. Kanbr O, Wali S, Wasfi L, et al. Prevalence and characteristics of rapid eye movement-related sleep apnea in a population-based cohort. Ann Thorac Med. 2025;20(2):117–124. doi:10.4103/atm.atm_188_24

    33. Berry RB, Brooks R, Gamaldo CE, et al. The AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications, Version 2.1. Darien, Illinois: American Academy of Sleep Medicine; 2014.

    34. AASM. American Academy of Sleep Medicine (AASM). International Classification of Sleep Disorders (ICSD). 3rd ed. Darien, IL: AASM; 2014.

    35. Conwell W, Patel B, Doeing D, et al. Prevalence, clinical features, and CPAP adherence in REM-related sleep-disordered breathing: a cross-sectional analysis of a large clinical population. Sleep Breath. 2012;16(2):519–526. doi:10.1007/s11325-011-0537-6

    36. Sakao S, Sakurai T, Yahaba M, et al. Features of REM-related sleep disordered breathing in the Japanese population. Intern Med. 2015;54(12):1481–1487. doi:10.2169/internalmedicine.54.4248

    37. Peduzzi P, Concato J, Kemper E, Holford TR, Feinstein AR. A simulation study of the number of events per variable in logistic regression analysis. J Clin Epidemiol. 1996;49(12):1373–1379. doi:10.1016/S0895-4356(96)00236-3

    38. Vittinghoff E, McCulloch CE. Relaxing the rule of ten events per variable in logistic and cox regression. Am J Epidemiol. 2007;165(6):710–718. doi:10.1093/aje/kwk052

    39. Faul F, Erdfelder E, Buchner A, Lang A-G. Statistical power analyses using G*power 3.1: tests for correlation and regression analyses. Behavior Research Methods. 2009;41(4):1149–1160. doi:10.3758/BRM.41.4.1149

    40. Cohen J. Statistical Power Analysis for the Behavioral Sciences (2nd Ed.). Hillsdale, NJ: Lawrence Erlbaum Associates; 1988.

    41. Brown LD, Cai TT, DasGupta A. Interval estimation for a binomial proportion. Stat Sci. 2001;16(2):101–133,133. doi:10.1214/ss/1009213286

    42. Newcombe RG. Two-sided confidence intervals for the single proportion: comparison of seven methods. Stat Med. 1998;17(8):857–872. doi:10.1002/(SICI)1097-0258(19980430)17:8<857::AID-SIM777>3.0.CO;2-E

    43. Pizza F, Tartarotti S, Poryazova R, Baumann CR, Bassetti CL. Sleep-disordered breathing and periodic limb movements in narcolepsy with cataplexy: a systematic analysis of 35 consecutive patients. Eur Neurol. 2013;70(1–2):22–26. doi:10.1159/000348719

    44. Frauscher B, Ehrmann L, Mitterling T, et al. Delayed diagnosis, range of severity, and multiple sleep comorbidities: a clinical and polysomnographic analysis of 100 patients of the innsbruck narcolepsy cohort. J Clin Sleep Med. 2013;9(8):805–812. doi:10.5664/jcsm.2926

    45. BaHammam AS, Alenezi AM. Narcolepsy in Saudi Arabia. Demographic and clinical perspective of an under-recognized disorder. Saudi Med J. 2006;27(9):1352–1357.

    46. Black J, Reaven NL, Funk SE, et al. Medical comorbidity in narcolepsy: findings from the burden of narcolepsy disease (BOND) study. Sleep Med. 2017;33:13–18. doi:10.1016/j.sleep.2016.04.004

    47. Maski K, Mignot E, Plazzi G, Dauvilliers Y. Disrupted nighttime sleep and sleep instability in narcolepsy. J Clin Sleep Med. 2022;18(1):289–304. doi:10.5664/jcsm.9638

    48. Terada J, Nakamura A, Zhang W, et al. Ventilatory long-term facilitation in mice can be observed during both sleep and wake periods and depends on orexin. J Appl Physiol. 2008;104(2):499–507. doi:10.1152/japplphysiol.00919.2007

    49. Zhang GH, Liu ZL, Zhang BJ, et al. Orexin A activates hypoglossal motoneurons and enhances genioglossus muscle activity in rats. Br J Pharmacol. 2014;171(18):4233–4246. doi:10.1111/bph.12784

    50. Yamada R, Koike T, Nakakariya M, Kimura H. Orexin receptor 2 agonist activates diaphragm and genioglossus muscle through stimulating inspiratory neurons in the pre-Bötzinger complex, and phrenic and hypoglossal motoneurons in rodents. PLoS One. 2024;19(6):e0306099. doi:10.1371/journal.pone.0306099

    51. Bonsignore MR, Mazzuca E, Baiamonte P, Bouckaert B, Verbeke W, Pevernagie DA. REM sleep obstructive sleep apnoea. Eur Respir Rev. 2024;33(171):230166. doi:10.1183/16000617.0166-2023

    52. Vilela M, Tracey B, Volfson D, et al. Identifying time-resolved features of nocturnal sleep characteristics of narcolepsy using machine learning. J Sleep Res. 2024;33(6):e14216. doi:10.1111/jsr.14216

    53. Hoshino T, Sasanabe R, Murotani K, et al. Estimated respiratory arousal threshold in patients with rapid eye movement obstructive sleep apnea. Sleep Breath. 2022;26(1):347–353. doi:10.1007/s11325-021-02399-9

    54. Zhou R, Suo C, Jiang Y, et al. Association of sleep pattern and genetic susceptibility with obstructive sleep apnea: a prospective analysis of the UK biobank. Nat Sci Sleep. 2024;16:503–515. doi:10.2147/NSS.S443721

    55. Huang EI, Huang SY, Lin YC, et al. Respiratory arousals in patients with very severe obstructive sleep apnea and how they change after a non-framework surgery. Healthcare. 2022;10(5). doi:10.3390/healthcare10050902

    56. Suzuki M, Shimamoto K, Sekiguchi H, et al. Arousal index as a marker of carotid artery atherosclerosis in patients with obstructive sleep apnea syndrome. Sleep Breath. 2019;23(1):87–94. doi:10.1007/s11325-018-1664-0

    57. Mokhlesi B, Varga AW. Obstructive sleep apnea and cardiovascular disease. rem sleep matters! Am J Respir Crit Care Med. 2018;197(5):554–556. doi:10.1164/rccm.201710-2147ED

    58. Kwon Y, Gami AS, Javaheri S, et al. Cardiovascular risks in people with narcolepsy: expert panel consensus recommendations. J Am Heart Assoc. 2024;13(16):e035168. doi:10.1161/JAHA.124.035168

    59. BaHammam AS. Cardiovascular disease risk and comorbidities in narcolepsy. In: BaHammam AS, Sharafkhaneh A, Pandi-Perumal SR, editors. Narcolepsy: Integrating Basic and Clinical Knowledge. London, United Kingdom: Academic Press, Elsevier; 2025:221–234.

    60. Eckert DJ. Phenotypic approaches to obstructive sleep apnoea – new pathways for targeted therapy. Sleep Med Rev. 2018;37:45–59. doi:10.1016/j.smrv.2016.12.003

    61. Sullivan SS, Guilleminault C. Emerging drugs for common conditions of sleepiness: obstructive sleep apnea and narcolepsy. Expert Opin Emerg Drugs. 2015;20(4):571–582. doi:10.1517/14728214.2015.1115480

    62. Mallampalli A. More than just narcolepsy: the importance of recognizing concurrent sleep disorders. In: BaHammam AS, Sharafkhaneh A, Pandi-Perumal SR, editors. Narcolepsy: Integrating Basic and Clinical Knowledge. Chennai, India: Elseveir; 2025:235–249.

    63. Zinchuk A, Yaggi HK. Phenotypic subtypes of OSA: a challenge and opportunity for precision medicine. Chest. 2020;157(2):403–420. doi:10.1016/j.chest.2019.09.002

    Continue Reading

  • Indonesia’s Janice Tjen on her US Open run and being inspired by Barty

    Indonesia’s Janice Tjen on her US Open run and being inspired by Barty

    Of the 12 players who successfully made it through US Open qualifying last week, Indonesia’s Janice Tjen garnered some of the most attention.

    The 23-year-old’s rise since graduating from Pepperdine University with a degree in sociology last year had been meteoric. Unranked last May, Tjen won 100 out of 113 matches over the next 16 months to rise to No. 149 this week, sweeping up 13 ITF titles in the process. But her opener against No. 25 seed Veronika Kudermetova represented another step up in level. It was not only Tjen’s tour-level debut, but the first time she’d ever faced a Top 50 opponent.

    US Open: Draws | Scores | Order of play

    Tjen responded brilliantly, upsetting Kudermetova 6-4, 4-6, 6-4 despite losing a break lead in the second set, and setting up a popcorn second-round encounter with Emma Raducanu. The seventh Indonesian woman to compete in a Grand Slam main draw in the Open Era, Tjen also snapped a long drought for her country at this level of the sport, becoming:

    • The first Indonesian woman to win a Grand Slam match since Angelique Widjaja at Roland Garros 2023
    • The first Indonesian woman to defeat a Top 30 player since Widjaja’s defeat of Patty Schnyder at Indian Wells 2003
    • The first Indonesian woman to win a US Open main-draw match since Widjaja’s defeat of Anna Kournikova in the 2002 first round
    • If Tjen beats Raducanu, she will be the first Indonesian woman to reach the third round of a major since former No. 19 Yayuk Basuki at Wimbledon 2000.

    I feel proud to be able to do this for my country,” Tjen told press afterwards. “Hopefully like this, by me making appearance here, will inspire more tennis player — like, younger kids to play tennis and also believing that they can be here too.”

    Growing up in Jakarta, Tjen got her start in tennis by accident. Her friend, Priska Nugroho — then one of the country’s top juniors, and the 2020 Australian Open girls’ doubles champion with Alexandra Eala — persuaded her to try the sport. Tjen’s parents quickly acquiesced.

    My parents were just like, ‘Yeah, just go play the sport, just having exercise,’ so I’m not going to be in my room just doing nothing,” Tjen recalled with a smile.

    Tjen patterned her game after a former World No. 1

    Against Kudermetova, Tjen excelled with a throwback game style that’s become all too rare in tennis: sliced backhands, a powerful forehand with which she struck a slew of lethal angled winners and an eagerness to get to the net, where she won 20 out of 30 points.

    “Me and my coach have been working on that,” Tjen said with satisfaction afterwards. “Just making sure that if it is a 50/50, we’re going in, coming into the net. And I would say we have a pretty good percentage of winning up at the net, so we would want to make more appearance up there.”

    It’s all reminiscent of none other than former World No. 1 Ashleigh Barty, who retired in 2022 — and it’s no surprise that Tjen has consciously patterned her game after the three-time major champion.

    “I have been hearing that a lot,” Tjen said. “She’s, I would say, my role model. I would watch a little bit of her matches and try to copy what works for me, what’s not, and just trying to understand her game a little bit more.”

    Tjen and Eala are both making history for south-east Asia in New York

    On a strong day for south-east Asian tennis, Tjen’s breakthrough was mirrored by none other than her former junior rival Eala, who delivered Day 1’s only other seeded upset, knocking out No. 14 Clara Tauson in a Grandstand barnburner. Eala, the first Filipina to win a Grand Slam main-draw match in the Open Era, remembered Tjen well from their junior rivalry.

    “Janice is super nice,” the 20-year-old told press. “I’ve known her for quite a long time. Growing up in the same region, we would run into each other a lot in the same tournaments. You know, I haven’t been able to spend a lot of time with her recently. I know she was in college. I’m so happy for her, and it’s nice to see someone that you grew up with in the biggest stages in the world.”

    Eala had the narrow edge in their 2018-19 junior days, leading their head-to-head at that level 2-1.

    Tjen was encouraged by her college coaches and a fellow Indonesian player

    Going pro was never an automatic decision for Tjen. After finishing with juniors, the travel costs involved meant that she and her parents opted for the college tennis pathway — which she feels paid off after her coaches at Pepperdine University helped to develop her game. And after spending her college career contemplating whether or not she should turn pro, they were also instrumental in encouraging her.

    “The coaches at Pepperdine told me I think you should give it a try, at least for two years,” she said. “So I trust them, and here I am!”

    The only other Indonesian player at the US Open — doubles No. 48 Aldila Sutjiadi — was also a key figure in Tjen’s emergence.

    “I am really, really close with Aldila,” Tjen said. “She’s always been like a good older sister to me, taking care of me, guiding me, and telling me, ‘This is what you need to do.’ She’s also one of the people that convinced me that I should give it a try. It’s very nice to be around another Indonesian here.”

    In her spare time, Tjen is a Mario Kart fan

    Tennis isn’t Tjen’s only competitive endeavor. In her downtime, she and her coach continue to battle hard — playing Mario Kart.

    “We’ve been grinding on that one,” she told press.

    Only one on the video game’s characters will do for Tjen, though — the green dinosaur Yoshi.

    “I’ve been into Mario Kart since I was reall young, and it’s always my go-to character,” she said. “I would be a little bit not happy if somebody took that. I would be like, ‘Nope, that’s my character.’”

     

    Continue Reading

  • Dengue cases surge in Khyber Pakhtunkhwa

    Dengue cases surge in Khyber Pakhtunkhwa

    – Advertisement –

    PESHAWAR, Aug 25 (APP):The spread of dengue virus in Khyber Pakhtunkhwa has reached an alarming level as the Health Department confirmed that the number of active cases has risen to 90 while 82 new patients were reported during the last 24 hours.

     According to the latest data issued by the Health Department, a total of 398 people have so far been infected with dengue in the province out of which 308 have completely recovered. Charsadda remains the most affected district where 73 active cases have been reported making it the hotspot of dengue spread in Khyber Pakhtunkhwa.

    Other districts have also reported cases including Abbottabad with 2 active patients, Haripur 1, Nowshera and Mansehra 3 each, Lakki Marwat 2, Mardan and Peshawar 1 each, Swabi 3 and Tank 1 case.

    The Health Department has advised the public to strictly follow precautionary measures, ensure cleanliness, eliminate stagnant water and use mosquito nets and repellents to prevent the spread of dengue. Citizens have also been urged to immediately approach the nearest hospital in case of any suspected symptoms.

    Continue Reading

  • MiR-491-5p Targets B4GalT5 to Alleviate Airway Inflammation and Remode

    MiR-491-5p Targets B4GalT5 to Alleviate Airway Inflammation and Remode

    Introduction

    Asthma is a common chronic inflammatory airway disease affecting over 300 million people worldwide, with increasing prevalence and significant clinical burden.1 Persistent airway inflammation and structural remodeling, especially the thickening and hyperplasia of airway smooth muscle (ASM), are central to disease progression and treatment resistance.2 Abnormal proliferation of ASM cells not only contributes to airway narrowing but also worsens airflow limitation and clinical outcomes.

    Recent studies highlight mitochondrial oxidative stress as a key driver of ASM dysfunction in asthma. Excessive reactive oxygen species (ROS) disrupt mitochondrial homeostasis, leading to impaired ATP synthesis, aberrant calcium signaling, and sustained inflammation.3–5 These mitochondrial alterations reinforce a vicious cycle of “inflammation-oxidative stress” in airway smooth muscle cells (ASMCs) that exacerbates airway inflammation and remodeling, underscoring the urgent need for novel molecular targets to interrupt this process.

    MicroRNAs (miRNAs) are key post-transcriptional regulators of gene expression and are implicated in the pathogenesis of numerous diseases. Among them, miR-491-5p has been reported to suppress cell proliferation, oxidative stress, and inflammation in cancer models.6–9 However, its role in chronic airway diseases such as asthma remains unexplored. This study aims to provide preliminary evidence regarding the potential role of miR-491-5p in the pathogenesis of asthma.

    To further elucidate the mechanism by which miR-491-5p functions in asthma, we used bioinformatics analysis to identify β-1,4-galactosyltransferase 5 (B4GalT5), a β-1,4-galactosyltransferase involved in oxidative stress pathways, as a direct target of miR-491-5p.10,11 Although B4GalT5 has been primarily studied in the context of tumor progression and cardiac hypertrophy, where it interacts with UGCG to promote ROS production and pathological remodeling,12 its role in asthma has not been explored. The involvement of B4GalT5 in airway inflammation and structural remodeling has yet to be elucidated.

    In this study, we systematically investigated the expression patterns and functional roles of miR-491-5p and B4GalT5 in asthma-related airway pathology. We found that miR-491-5p was downregulated, while B4GalT5 was upregulated in ASM tissues of asthma patients. Functionally, miR-491-5p overexpression inhibited TNF-α-induced ASMC proliferation, inflammation, and mitochondrial oxidative stress, whereas B4GalT5 overexpression reversed these effects. These findings suggest that miR-491-5p may alleviate airway inflammation and remodeling in asthma by targeting B4GalT5, providing novel insights into the asthma-related molecular mechanisms and future treatment strategies.

    Materials and Methods

    Ethics Statement

    The collection and use of human specimens in this study were approved by the Medical Ethics Committee of the People’s Hospital of Zhengzhou University (Approval No.202407701). All procedures strictly adhered to the ethical principles outlined in the Declaration of Helsinki. Written informed consent was obtained from all participants. The privacy and confidentiality of all subjects were strictly protected throughout the study.

    Human Study

    Inclusion criteria were as follows: (1) diagnosis of bronchial asthma based on the Global Initiative for Asthma (GINA) guidelines;13 (2) age between 18 and 65 years; (3) surgical specimens containing structurally intact segmental bronchi; (4) no acute asthma exacerbation within the past 4 weeks; (5) no systemic corticosteroid treatment within 4 weeks prior to surgery. Exclusion criteria included: (1) presence of other respiratory diseases besides asthma, such as chronic obstructive pulmonary disease (COPD), pulmonary hypertension, or lung cancer; (2) systemic inflammatory or autoimmune diseases such as systemic lupus erythematosus or rheumatoid arthritis; (3) severe cardiovascular, cerebrovascular, or other systemic diseases affecting lung function. Patients undergoing surgery for pulmonary nodules were used as the normal control group. All control subjects had normal pulmonary function and no clinical or histological evidence of airway inflammation. ASM tissues were collected from segmental or subsegmental bronchi in lung regions anatomically distant from the pulmonary nodules. These bronchi were histologically normal and clearly surrounded by smooth muscle layers, ensuring the reliability of the ASM samples as representative of healthy airways. Pulmonary function was assessed using the MasterScreen® spirometry system (Viasys Healthcare GmbH, Hoechberg, Germany).

    Human Specimens

    Segmental bronchial tissues resected during surgery were immediately transported on ice to the laboratory. After thorough rinsing with phosphate-buffered saline (PBS, Servicebio, Wuhan, China) to remove surface blood and mucus, segmental bronchi were dissected under a stereomicroscope. The bronchi were longitudinally opened, and the mucosal and adventitial layers were removed, retaining only the central smooth muscle layer. Residual glandular and cartilaginous structures were carefully scraped off to isolate purified airway smooth muscle tissue. Remaining tissues were fixed in 4% paraformaldehyde and embedded in paraffin for subsequent histological staining and analysis.

    HRCT Scanning

    All participants underwent high-resolution computed tomography (HRCT, Philips, Netherlands) of the chest at the end of a quiet inspiratory phase. The scanning parameters were as follows: 1 mm slice thickness, 1 mm interslice gap, 120 kVp tube voltage, and 100–200 mA tube current. Subjects were scanned in the supine position from the lung apex to the base. Images were imported into the Carestream Health system for 3D reconstruction and analysis. To reduce inter-individual anatomical variation, we standardized our measurements by consistently selecting the apical segmental bronchus of the right lower lobe (RB10) in all subjects and performing all assessments at the same anatomical level across participants. Cross-sectional areas of the airway were manually or semi-automatically outlined to measure airway diameter (D) and luminal diameter (L) (Supplementary Figure S1). The percentage of wall area (WA%) was calculated using the following formula:


    Two radiologists independently measured the images in a double-blinded manner.

    miRNA Sequencing and Analysis

    After dissection of pure airway smooth muscle tissues from segmental bronchi of asthma patients (n=3) and control participants (n=3), total RNA was immediately extracted using Trizol reagent (Thermo Fisher, Waltham, MA, USA). The RNA concentration and purity were assessed using a NanoDrop 2000 spectrophotometer (Thermo Fisher), ensuring an OD260/280 ratio between 1.8 and 2.0. RNA integrity was evaluated using the Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA), and only samples with an RNA integrity number (RIN) ≥7 were used for library construction. Qualified RNA samples were shipped on dry ice to Aiji Baike Biotechnology Co., Ltd. (Wuhan, Hubei, China) for small RNA library preparation and high-throughput sequencing. The library preparation included 3′ and 5′ adaptor ligation, reverse transcription, qRT-PCR amplification, and target fragment selection. Single-end sequencing was performed using the Illumina platform. Raw reads were subjected to quality control, including adaptor trimming and removal of low-quality reads, to obtain clean reads of 18–30 nt in length. Clean reads were mapped to the miRBase database (v22) to annotate known miRNAs. Differential expression analysis was conducted using DESeq2, with screening criteria of |log2FoldChange| > 1 and P < 0.05.

    Animals and Treatments

    Female C57BL/6 mice (20–22 g) were purchased from SPF Biotechnology Co., Ltd. (Beijing, China). Mice were housed under specific pathogen-free (SPF) conditions with a temperature of 22 ± 2°C, relative humidity of 45–55%, and a 12-hour light/dark cycle. Mice were randomly assigned to four groups (n=6/group): (A) Control; (B) Asthma; (C) Asthma + AAV-NC-miR-491-5p; (D) Asthma + AAV-miR-491-5p.AAV-NC and AAV-miR-491-5p vectors (Fenghui Biotechnology Co., Ltd., Changsha, Hunan, China) were intratracheally instilled (50 μL per mouse) on days 0, 7, and 15. On days 0, 7, and 14, mice in groups B, C, and D were sensitized by intraperitoneal injection of 40 μg ovalbumin (OVA; Yuanye, Shanghai, China) mixed with 2 mg aluminum hydroxide in 0.2 mL saline. From day 21, these groups were challenged with 5% OVA aerosol by ultrasonic nebulization for 30 minutes per day for 8 consecutive days. Control mice received equal volumes of saline at all time points. No adverse effects were observed in any mice treated with AAV vectors, and all procedures were well tolerated. Mice were euthanized on day 30 with an intraperitoneal dose of 200 mg/kg sodium pentobarbital. The experimental protocol was approved by the Medical Ethics Committee of Zhengzhou University (Approval No. ZZU-LAC20240628[02]) and complied with the Helsinki Declaration and relevant guidelines for animal welfare.

    Sample Collection

    On day 30, mice were euthanized, and the trachea was exposed under sterile conditions. Bronchoalveolar lavage fluid (BALF) was collected by slowly instilling 1 mL sterile PBS (containing 1% protease inhibitor) in three aliquots of 0.3 mL, retaining each for 10 seconds before withdrawal. The recovery rate was >80%. The lavage fluid was centrifuged at 300 ×g for 10 minutes at 4°C, and the supernatant was stored at –80°C for subsequent analysis, while cell pellets were used for inflammatory cell counts. Approximately 0.8–1 mL of whole blood was drawn from the left ventricle using a 1 mL syringe, transferred into EDTA anticoagulation tubes, gently mixed, and stored at 4°C. Lungs were perfused with pre-cooled PBS via the pulmonary artery until the tissue turned white. The left lung was fixed in 4% paraformaldehyde, and the right lung was snap-frozen at –80°C.

    Histologic Analysis of Lung Tissue

    Lung tissues were fixed in 4% paraformaldehyde for 24 hours, then dehydrated, embedded in paraffin, and sectioned at 5 μM for histological staining and morphological evaluation. Paraffin sections were deparaffinized, rehydrated, and stained with Hematoxylin and Eosin (HE; Baso, Guangzhou, China), Periodic Acid–Schiff (PAS; Solarbio, Beijing, China), and Masson trichrome stain (Solarbio) to assess inflammatory infiltration, goblet cell hyperplasia, mucus secretion, and collagen deposition. For immunohistochemistry, deparaffinized sections were subjected to antigen retrieval in citrate buffer (pH 6.0) at 121°C for 2 minutes. After PBS washing, sections were incubated overnight at 4°C with anti-B4GalT5 (Abmart, Shanghai, China) and anti-Ki-67 (Abclone, Wuhan, China) antibodies. After washing, HRP-conjugated anti-rabbit IgG (Thermo Fisher) was added and incubated at room temperature for 1 hour, followed by DAB chromogenic reaction (Solarbio), PBS rinse, hematoxylin counterstaining, and neutral resin mounting. Images were captured using a CX21 light microscope (Olympus, Tokyo, Japan), and quantitative scoring was performed. Inflammatory scoring (0–4) was based on HE staining: 0, no inflammation; 1, mild peribronchial infiltration; 2, moderate infiltration involving airways and vessels; 3, large clusters or layered distribution; 4, extensive infiltration with architectural changes.

    PAS staining was scored from 0 to 6 based on goblet cell hyperplasia (0–3) and mucus production (0–3). Goblet cell score: 0, none; 1, 15–30% epithelial cells; 2, 30–50%; 3, >50%. Mucus production score: 0, none; 1, <1/3 airway circumference; 2, 1/3–2/3; 3, complete obstruction. Collagen deposition was assessed by measuring the percentage of blue collagen fiber area around the airway using ImageJ (v2.14.0). HE-stained images were used to quantify airway remodeling. Airways with 100–200 μM luminal diameter, complete and nearly circular, were selected at 20× magnification. For each mouse, three fields were randomly selected and averaged. As previously reported,14 the following parameters were measured using ImageJ: internal perimeter (Pi), wall area (WAi), smooth muscle area (WAm), and number of smooth muscle cells (N). Normalized indicators—WAi/Pi, WAm/Pi, WAi/WAm and N/Pi—were calculated to evaluate wall thickening and ASM remodeling. All experiments were performed in triplicate, and results were averaged for statistical analysis.

    Cell Culture and Transfection

    Airway smooth muscle cells were isolated from 6-week-old C57BL/6 mice and cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin at 37°C in 5% CO₂. 293T cells were purchased from Fenghui Biotechnology Co., Ltd. and cultured in DMEM with 10% FBS. For TNF-α stimulation experiments, ASMCs were transfected with miR-491-5p mimic, miR-491-5p-NC, pcDNA3.1-B4GalT5, or pcDNA3.1-NC (Fenghui Biotechnology Co., Ltd.) using Lipofectamine 3000 (Thermo Fisher) for 24 hours, followed by treatment with 25 ng/μL TNF-α (Beyotime) for 24 hours. The cells were then collected for further analysis.

    Quantitative Real-Time PCR (qRT-PCR)

    Total RNA was extracted from cells or tissues using Trizol (Thermo Fisher) or the miRNA Purification Kit (Yeasen, Shanghai, China). Reverse transcription was performed using the 1st Strand cDNA Synthesis SuperMix (Yeasen) for mRNA and the miRNA 1st Strand cDNA Synthesis Kit for miRNA, according to the manufacturer’s instructions, with 1 μg of total RNA or miRNA. qRT-PCR was conducted using SYBR Green Master Mix (Yeasen), and specific primers were used to amplify target genes and miRNAs. Each sample was analyzed in triplicate. Relative expression levels were calculated using the 2−ΔΔCt method, with GAPDH or U6 used as internal controls. Specific primer sequences are detailed in Supplementary Table S1.

    Western Blot

    Total protein was extracted from cells and tissues using RIPA lysis buffer supplemented with 100 μg/mL PMSF and protease inhibitors. Protein concentrations were determined using the BCA Protein Assay Kit (Beyotime). Samples were mixed with 5× reducing loading buffer at a ratio of 4:1 and denatured at 100°C for 10 minutes. Equal amounts of protein (20–40 μg) were separated via SDS-PAGE and transferred onto PVDF membranes (Millipore, Bedford, MA, USA) using wet transfer. Membranes were blocked with 5% non-fat milk at room temperature for 1 hour and incubated overnight at 4°C with primary antibody against B4GalT5 (1:1000, Abmart, Shanghai, China). After washing with TBST, membranes were incubated with Goat anti-Rabbit secondary antibody (1:800, LI-COR, Lincoln, Nebraska, USA) in the dark at room temperature for 1 hour. Blots were visualized using the Li-COR ODYSSEY9120 imaging system. Band intensity was quantified using ImageJ software, and GAPDH (1:2000, Abmart) was used as the loading control.

    Dual-Luciferase Reporter Assay

    The wild-type (WT) and mutant (MUT) 3′-UTR fragments of B4GalT5 were synthesized and cloned into the psi-CHECK-2 vector, designated as B4GalT5-WT and B4GalT5-MUT, respectively. These constructs were co-transfected with miR-491-5p mimics or negative control (NC) into HEK293T cells using Lipofectamine 3000 (Thermo Fisher). After 48 hours of transfection, luciferase activity was measured using the Dual-Luciferase Reporter Assay System (Gene Creat, Wuhan, Hubei, China).

    ELISA

    The levels of IL-6, IL-8, and IL-1β in cell culture supernatants and BALF were measured using ELISA kits (Coibo, Shanghai, China) according to the manufacturer’s instructions. The detection sensitivity for IL-6, IL-8, and IL-1β was 0.1 pg/mL.

    Cell Counting Kit (CCK)-8 Assay

    Cell proliferation was assessed using the CCK-8 kit (Mlbio, Shanghai, China) following the manufacturer’s instructions. Briefly, cells were seeded at 5×10³ cells per well in a 96-well plate and incubated overnight. At the indicated time points (24, 48, and 72 hours), 10 μL of CCK-8 solution was added to each well and incubated at 37°C for 2 hours. Absorbance was measured at 450 nm using a microplate reader (BECKman Coulter, California, USA).

    Measurement of Total Reactive Oxygen Species (ROS)

    ASMCs were washed three times with PBS and incubated with 10 μM DCFH-DA (UElandy, Suzhou, Jiangsu, China) at 37°C in the dark for 20–30 minutes. After incubation, cells were washed with PBS three times to remove excess probe. Green fluorescence (indicative of ROS levels) was observed and imaged using a fluorescence microscope (FITC channel, excitation 488 nm, emission 525 nm). For tissue detection, fresh lung tissues were sectioned into 8 μM frozen slices, washed with PBS, incubated with 10 μM DCFH-DA for 30 minutes, then washed and imaged. The mean gray value of fluorescence images was quantified using ImageJ software. Multiple fields per sample were analyzed for representative and accurate quantification.

    Malondialdehyde (MDA) Assay

    MDA levels in lung tissues were measured using the MDA Assay Kit (Beyotime). Lung tissues were finely minced and homogenized with physiological saline on ice. Homogenates were centrifuged at 12,000 rpm for 10 minutes at 4°C, and the supernatants were collected. According to the manufacturer’s instructions, the supernatant was incubated with MDA reaction reagents at 37°C for 30 minutes. Absorbance was measured at 532 nm using a spectrophotometer or microplate reader, and MDA concentration was calculated based on a standard curve, expressed as μmol/g tissue.

    Superoxide Dismutase (SOD) Activity Assay

    Supernatants from 10% lung tissue homogenates (prepared as described in the MDA assay) were used to measure SOD activity using the SOD Assay Kit (Beyotime). Reactions were carried out in 96-well plates, and absorbance was measured at 450 nm. SOD activity was calculated using a standard curve and expressed as U/mg protein.

    Adenosine Triphosphate (ATP) Level Detection

    Lung tissues were homogenized in pre-chilled lysis buffer on ice and centrifuged at 12,000 rpm for 10 minutes at 4°C to collect the supernatant. According to the ATP Assay Kit (Beyotime), 10 μL of the supernatant was mixed with 100 μL of ATP reaction solution containing a fluorescence substrate. Fluorescence intensity was measured at 485 nm using a fluorescence microplate reader. ATP levels were expressed as nmol/mg protein.

    Transmission Electron Microscopy (TEM)

    ASMCs were washed 2–3 times with pre-cooled PBS and fixed with 2.5% glutaraldehyde (Solarbio) at 4°C for 6 hours. Samples were washed three times with PBS (10 minutes each), dehydrated using graded ethanol solutions (50%, 70%, 80%, 90%, 95%, 100%, 10 minutes each), followed by acetone replacement. Samples were embedded in Epon812 (Solarbio) and polymerized at 60°C for 48 hours. Ultrathin sections (~70 nm) were cut, placed on copper grids, and stained with uranyl acetate (15 minutes) and lead citrate (10 minutes) (Solarbio). Sections were air-dried and observed under a transmission electron microscope to visualize mitochondrial morphology.

    Quantitative analysis of mitochondrial morphology was performed using TEM images. Damaged mitochondria were defined as those exhibiting at least two of the following features: disrupted or lost cristae, mitochondrial swelling, vacuolization, or irregular shape. For each sample, 5–10 random fields were analyzed, and at least 50 mitochondria were evaluated per group. The percentage of damaged mitochondria was calculated as the ratio of damaged mitochondria to the total mitochondria. All quantifications were conducted in a blinded manner by two independent observers.

    Intracellular Calcium Ion (Ca2+) Detection

    After treatment, cells were washed twice with pre-warmed PBS and incubated with 5 μM Fluo-4 AM (Beyotime) dissolved in HBSS containing 0.02% Pluronic F-127 at 37°C for 30 minutes to allow intracellular loading. After incubation, cells were washed three times with PBS to remove excess dye. Green fluorescence (indicating intracellular calcium levels) was detected by flow cytometry (BD Biosciences, FACSCalibur) under 494 nm excitation. Fluorescence intensity was analyzed using FlowJo (v10.8.1), and mean fluorescence intensity was used to reflect intracellular calcium levels.

    Statistical Analysis

    All data represent the average of at least three separate experiments. Statistical analyses were performed using GraphPad Prism (v10.3.1). Data are presented as mean ± standard error of the mean (Mean ± SEM). Comparisons between two groups were conducted using unpaired Student’s t-test, while comparisons among multiple groups were analyzed using one-way ANOVA followed by Tukey’s post hoc test. A P-value < 0.05 was considered statistically significant (*P<0.05, **P<0.01, ***P<0.001, ****P<0.0001).

    Results

    Downregulation of miR-491-5p and Upregulation of B4GalT5 in Airway Smooth Muscle of Asthma Patients

    To investigate the role of miRNAs in asthma, we collected segmental bronchial tissues from three asthma patients and three control participants. The ASM was isolated and subjected to miRNA-Seq. A total of 22 differentially expressed miRNAs were identified, among which miR-491-5p was the only significantly downregulated miRNA. This unique expression pattern prompted us to focus on miR-491-5p for subsequent studies (Figure 1a). To systematically explore the regulatory network of miR-491-5p, we predicted its downstream target genes using three databases: TargetScan, miRDB, and miRTarBase. After merging the results and removing duplicates, eight candidate genes were identified (Figure 1b). Among them, B4GalT5 emerged as a key focus of this study due to its well-documented role in regulating oxidative stress and its scarce reporting in asthma research. Using the TargetScan Human 8.0 tool, we predicted a potential binding site between miR-491-5p and the 3’UTR of B4GalT5, revealing a sequence of seven consecutive complementary bases, indicating a potentially stable interaction (Figure 1c). To further validate this regulatory relationship, we analyzed miRNA and mRNA expression data of ASMCs from asthma patients using the GEO dataset GSE119580. The results showed a significant negative correlation between miR-491-5p and B4GalT5 expression (Figure 1d). Based on these findings, we further explored the expression patterns and functional implications of miR-491-5p and B4GalT5 in the ASM of asthma patients.

    Figure 1 Screening and target prediction of miR-491-5p in asthma airway smooth muscle (a) Volcano plot showing differentially expressed miRNAs identified by miRNA-Seq of airway smooth muscle from asthma patients (n=3) versus control participants (n=3). miR-491-5p was the only miRNA significantly downregulated (|log2FoldChange| > 1, adjusted P < 0.05, highlighted in red frame). (b) A total of 8 predicted downstream genes were screened through the database. B4GalT5, marked in red, was selected for further study. (c) Predicted binding sequence between B4GalT5 mRNA 5’UTR and miR-491-5p. (d) Negative correlation between miR-491-5p and B4GalT5 expression in airway smooth muscle transcriptome data. Correlations were assessed using Spearman’s rank correlation analysis (R² = 0.22, P < 0.05).

    To assess the expression of miR-491-5p and B4GalT5 in ASM and their potential roles, we performed qRT-PCR analysis on ASM from asthma patients and control participants. As expected, miR-491-5p expression was significantly downregulated, while B4GalT5 mRNA levels were markedly upregulated in the asthma group compared to control participants (Figure 2a and b). To further confirm the protein expression of B4GalT5, we conducted WB analysis, which consistently showed a significant increase in B4GalT5 protein levels in the ASM of asthma patients (Figure 2c). To investigate the potential role of B4GalT5 in ASM proliferation in asthma, we performed immunohistochemical staining to detect Ki-67 and B4GalT5 expression (Figure 2d and e). The result showed the expression level of B4GalT5 was positively correlated with the proportion of Ki-67–positive cells (Figure 2f). Since Ki-67 is a key marker of cell proliferation, its increased expression suggests that B4GalT5 may be involved in regulating ASM proliferation in asthma. Furthermore, we compared the airway wall area as a percentage of total airway area (WA%) between asthma patients and control participants, and found that WA% was significantly higher in the asthma group (Figure 2g and h). Additional analysis revealed a strong positive correlation between B4GalT5 expression and WA% (Figure 2i), supporting a link between B4GalT5 expression and structural airway remodeling, and providing further evidence for its potential role in ASM proliferation. Together, these results demonstrate that miR-491-5p is significantly downregulated, while B4GalT5 is significantly upregulated in the ASM of asthma patients. Moreover, miR-491-5p may participate in the regulation of ASM proliferation by negatively regulating B4GalT5.

    Figure 2 Altered expression of miR-491-5p and B4GalT5 associated with airway remodeling in asthma (a) The mRNA expression of miR-491-5p in airway smooth muscle were measured by qRT-PCR in both asthmatic patients (n=7) and control participants (n=3). (b) The mRNA expression of B4GalT5 in airway smooth muscle were measured by qRT-PCR in both asthmatic patients (n=7) and control participants (n=3). (c) Western blot analysis detected the protein expression of B4GalT5 in airway smooth muscle tissues from both asthmatic patients (n=7) and control participants (n=3). (d) The protein expression of B4GalT5 in airway smooth muscle were detected by IHC in both asthmatic patients (n=3) and control participants (n=3). Scale bar, 1 mm. (e) The protein expression of Ki-67 in airway smooth muscle were detected by IHC in both asthmatic patients (n=3) and control participants (n=3). Scale bar, 1 mm. (f) Correlation between B4GalT5 and Ki-67 expression levels in airway smooth muscle was assessed using Spearman analysis (Controls n=3; Asthma n=3). (g) Representative HRCT images of bronchial wall structure from control participants (n=3) and asthmatic patients (n=7). Red arrows point to the cross-section of the apex of the right lower lobe (RB10) bronchus, which was used to quantify WA%. (h) Measurement data of single-layer airway at the apex of the right lower lobe (RB10) (Controls n=3; Asthma n=7). (i) Correlation between B4GalT5 expression levels in airway smooth muscle and airway wall thickness in asthmatic patients was assessed using Spearman analysis. Data are presented as mean ± SEM and three or more independent experiments were performed. Group comparisons were performed using independent-samples t-test. Correlations were assessed using Spearman’s rank correlation analysis. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

    Abbreviations: qRT-PCR, quantitative reverse transcription polymerase chain reaction; IHC, immunohistochemistry; HRCT, high-resolution computed tomography.

    B4GalT5 Is a Direct Target of miR-491-5p

    To further confirm the targeting relationship between miR-491-5p and B4GalT5, as well as the predicted binding site, we performed a dual-luciferase reporter assay. The results showed that miR-491-5p significantly reduced the luciferase activity of the wild-type B4GalT5 3′UTR construct, whereas it had no effect on the mutant B4GalT5 3′UTR construct (Figure 3). These findings confirm that miR-491-5p directly binds to the 3′UTR of B4GalT5 mRNA, indicating a specific post-transcriptional regulatory relationship.

    Figure 3 B4GalT5 were target of miR-491-5p Dual-luciferase reporter assay confirmed the interaction between miR-491-5p and B4GalT5. ***P<0.001.

    Abbreviation: ns, not significant.

    Overexpression of miR-491-5p Suppresses TNF-α-Induced Proliferation, Inflammation, and Mitochondrial Oxidative Stress in Airway Smooth Muscle Cells by Targeting B4GalT5

    To investigate the role of miR-491-5p in the pathogenesis of asthma, ASMCs were treated with TNF-α to induce inflammatory responses and proliferation. Cells were divided into four groups: control, TNF-α, TNF-α + miR-491-5p mimic, and TNF-α + miR-491-5p NC. qRT-PCR and Western blot analyses revealed that miR-491-5p expression was significantly downregulated in the TNF-α group compared with the control group, while its target gene B4GalT5 was significantly upregulated at both mRNA and protein levels. Transfection with miR-491-5p mimic led to a significant increase in miR-491-5p expression, accompanied by a marked suppression of B4GalT5 expression at both the mRNA and protein levels, confirming successful transfection and target regulation (Figure 4a–c). Morphological observations and cell density quantification under a light microscope showed that, after 72 hours of TNF-α stimulation, ASMCs density was markedly increased, while miR-491-5p overexpression reduced this proliferative response (Figure 4d and e). Consistently, CCK-8 assays showed that TNF-α stimulation significantly promoted ASMCs viability, whereas miR-491-5p mimic treatment effectively attenuated TNF-α-induced proliferation (Figure 4f). ELISA analysis indicated that TNF-α stimulation markedly increased the levels of pro-inflammatory cytokines IL-6, IL-8, and IL-1β, while miR-491-5p overexpression significantly reduced their expression (Figure 4g).

    Figure 4 Effects of miR-491-5p on proliferation, inflammation, oxidative stress, and mitochondrial function in ASMCs (a) qRT-PCR verified the changes of miR-491-5p mRNA expression under TNF-α stimulation and after transfection of miR-491-5p mimic. (b) qRT-PCR verified the changes of B4GalT5 mRNA expression under TNF-α stimulation and after transfection of miR-491-5p mimic. (c) Western blot verified the changes of B4GalT5 protein expression under TNF-α stimulation and after transfection of miR-491-5p mimic. (d) The density and morphology of ASMCs. Scale bar, 100 μm. (e) Quantitative analysis of ASMCs cell density. (f) CCK-8 detected proliferation of ASMCs at indicated time (24, 48 or 72h). (g) ELISA assay examined the IL-6, IL-8 and IL-1β level in ASMCs. (h) DCFH-DA dye was used to detect the expression of ROS in ASMCs. Scale bar, 100 μm. (i) ROS quantification. (jk) Representative transmission electron microscopy images and quantitative analysis of mitochondrial morphology (arrowhead represents mitochondria). Scale bars: 2 μm (low magnification), 500 nm (high magnification). (l) Flow cytometry was performed to measure the mean fluorescence intensity of intracellular Ca²⁺ in ASMCs. (m) Ca2+ fluorescence intensity. Data are presented as means ± SEM and three or more independent experiments were performed. Significance was calculated by one-way ANOVA followed by Tukey’s post-hoc test. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

    Abbreviations: qRT-PCR, quantitative reverse transcription polymerase chain reaction; ASMCs, airway smooth muscle cells; ROS, reactive oxygen species.

    Furthermore, intracellular ROS levels were measured using the DCFH-DA fluorescent probe. DCFH-DA itself is non-fluorescent and becomes fluorescent DCF upon oxidation by ROS inside cells. Quantitative analysis of DCF fluorescence using ImageJ revealed that ROS levels were significantly elevated in the TNF-α group compared with the control group, while miR-491-5p overexpression significantly attenuated ROS accumulation (Figure 4h and i). Given that mitochondria are a major target of ROS-induced cellular damage, we further examined mitochondrial ultrastructure using transmission electron microscopy. In the control group, mitochondria displayed typical elongated or branched morphology with well-organized and intact structures. In contrast, TNF-α treatment significantly increased the proportion of damaged mitochondria and induced characteristic morphological alterations including swelling and vacuolization. Notably, overexpression of miR-491-5p markedly alleviated TNF-α-stimulated mitochondrial damage (Figure 4j and k). Considering that mitochondria are the main intracellular reservoirs of calcium and that calcium homeostasis is closely associated with mitochondrial oxidative stress, we further investigated whether miR-491-5p regulates intracellular calcium levels. Fluorescence imaging with a calcium-sensitive dye showed that TNF-α significantly elevated intracellular Ca²⁺ levels in ASMCs, whereas miR-491-5p overexpression significantly restored calcium homeostasis (Figure 4l and m).

    Overexpression of B4GalT5 Reverses the Inhibitory Effects of miR-491-5p on TNF-α-Induced Airway Smooth Muscle Cells Proliferation, Inflammation and Mitochondrial Oxidative Stress

    To further elucidate whether the functional regulation of ASMCs by miR-491-5p is mediated through B4GalT5, we first transfected ASMCs with plasmids overexpressing B4GalT5 or control vector. The overexpression efficiency of B4GalT5 was verified by both qRT-PCR and Western blot, which showed significantly increased B4GalT5 expression at the mRNA and protein levels in the pcDNA3.1-B4GalT5 group compared with the vector control (Figure 5a and b). These results confirm the effective transfection and functional overexpression of B4GalT5 in ASMCs. Subsequently, cells were divided into two groups for rescue experiments: TNF-α + miR-491-5p mimic + pcDNA3.1-vector group and TNF-α + miR-491-5p mimic + pcDNA3.1-B4GalT5 group. After 72 hours of treatment, cell morphology was examined under a light microscope, and cell density was quantified. The results showed that overexpression of B4GalT5 significantly reversed the inhibitory effect of miR-491-5p on TNF-α-induced ASMCs proliferation (Figure 5c and d). CCK-8 assays further confirmed that B4GalT5 overexpression significantly reversed the inhibitory effect of miR-491-5p on cell proliferation (Figure 5e). ELISA results indicated that the levels of inflammatory cytokines IL-6, IL-8, and IL-1β were significantly elevated in the TNF-α + miR-491-5p mimic + pcDNA3.1-B4GalT5 group compared to the TNF-α + miR-491-5p mimic + pcDNA3.1-vector group, suggesting that B4GalT5 can counteract the inhibitory effects of miR-491-5p on cytokine production in TNF-α-stimulated ASMCs (Figure 5f).

    Figure 5 B4GalT5 overexpression reverses the protective effects of miR-491-5p on proliferation, inflammation, and oxidative stress in ASMCs (a) qRT-PCR verified the changes of B4GalT5 mRNA expression in ASMCs following co-transfection with miR-491-5p mimic and pcDNA3.1-B4GalT5 under TNF-α stimulation. (b) Western blot validated the rescue of B4GalT5 protein expression in ASMCs transfected with miR-491-5p mimic following TNF-α stimulation. (c) The density and morphology of ASMCs. Scale bar, 100 μm. (d) Quantitative analysis of ASMCs cell density. (e) CCK-8 detected proliferation of ASMCs at indicated time (24, 48 or 72h). (f) ELISA assay examined the IL-6, IL-8 and IL-1β level in ASMCs. (g) DCFH-DA dye was used to detect the expression of ROS in ASMCs. Scale bar, 100 μm. (h) ROS quantification. (ij) Representative transmission electron microscopy images and quantitative analysis of mitochondrial morphology (arrowhead represents mitochondria). Scale bars: 2 μm (low magnification), 500 nm (high magnification). (k) Flow cytometry was performed to measure the mean fluorescence intensity of intracellular Ca²⁺ in ASMCs. (l) Ca2+ fluorescence intensity. Data are presented as means ± SEM and three or more independent experiments were performed. Group comparisons were performed using independent-samples t-test. *P<0.05, **P<0.01, ****P<0.0001.

    Abbreviations: qRT-PCR, quantitative reverse transcription polymerase chain reaction; ASMCs, airway smooth muscle cells; ROS, reactive oxygen species.

    To further explore the impact of B4GalT5 on mitochondrial oxidative stress, we measured intracellular ROS levels and observed mitochondrial ultrastructural changes. DCFH-DA fluorescence probe assays showed that ROS levels were significantly elevated in the TNF-α + miR-491-5p mimic + pcDNA3.1-B4GalT5 group compared to the TNF-α + miR-491-5p mimic + pcDNA3.1-vector group (Figure 5g and h), indicating that miR-491-5p inhibits TNF-α-induced oxidative stress, at least in part, by targeting B4GalT5. TEM further revealed that in the TNF-α + miR-491-5p mimic + pcDNA3.1-B4GalT5 group, mitochondria exhibited significant pathological changes, including matrix swelling and increased cristae spacing. Quantitative analysis showed a markedly increased proportion of damaged mitochondria in this group, indicating that excessive ROS accumulation induced by B4GalT5 overexpression leads to mitochondrial oxidative stress damage, thereby reversing the protective effects of miR-491-5p on mitochondrial morphology and function (Figure 5i and j). Furthermore, intracellular calcium levels were assessed using a calcium-sensitive fluorescent probe. The results showed a significant increase in Ca²⁺ fluorescence intensity in the TNF-α + miR-491-5p mimic + pcDNA3.1-B4GalT5 group compared to the TNF-α + miR-491-5p mimic + pcDNA3.1-vector group (Figure 5k and l), indicating that overexpression of B4GalT5 disrupts intracellular Ca²⁺ homeostasis. These findings suggest that B4GalT5 may exacerbate mitochondrial dysfunction by disturbing calcium homeostasis.

    Overexpression of miR-491-5p Alleviates Airway Remodeling, Pulmonary Inflammation, and Mitochondrial Oxidative Stress in Asthmatic Mice

    To investigate the regulatory role of miR-491-5p in airway remodeling, pulmonary inflammation, and mitochondrial oxidative stress in vivo, we established an ovalbumin (OVA)-induced asthma mouse model. 24 nude mice were randomly distributed across four groups: control, asthma, asthma + AAV-miR-491-5p, and asthma + AAV-NC-miR-491-5p. A typical allergic asthma mouse model was induced by OVA stimulation,15 and miR-491-5p-AAV was used for intervention treatment (Figure 6a). qRT-PCR results showed that, compared to the control group, miR-491-5p expression was significantly reduced in the airway smooth muscle tissue of the OVA group, while B4GalT5 expression was significantly increased. Importantly, in the asthma + AAV-miR-491-5p group, miR-491-5p expression was significantly upregulated compared to the asthma group, indicating successful in vivo delivery of miR-491-5p via AAV. Correspondingly, B4GalT5 expression was significantly downregulated in this group (Figure 6b and c). Western blot further confirmed that, compared to the control group, B4GalT5 protein levels were significantly elevated in the airway smooth muscle of the OVA group, and miR-491-5p-AAV intervention significantly reversed this phenomenon (Figure 6d). These results indicate that miR-491-5p can similarly target and inhibit B4GalT5 expression in vivo. We then measured the inner perimeter (Pi), internal area (WAi), smooth muscle area (WAm), and the number of bronchial smooth muscle cells (N). The results showed that, compared to the control group, the asthma group exhibited increased N/Pi, WAi/Pi, and WAm/Pi levels, while the Wai/WAm ratio decreased. However, miR-491-5p-AAV intervention significantly reversed these changes in the asthma mice (Figure 6e–h). These results indicate that miR-491-5p significantly improves airway remodeling in asthma mice.

    Figure 6 miR-491-5p attenuates airway inflammation and remodeling in OVA-induced asthma mouse model (a) Schematic diagram of OVA-induced asthma model establishment and AAV-miR-491-5p treatment. (b) qRT-PCR verified the changes of miR-491-5p mRNA expression in asthma model mice and with or without AAV-miR-491-5p treatment. (c) qRT-PCR verified the changes of B4GalT5 mRNA expression in asthma model mice and with or without AAV-miR-491-5p treatment. (d) Western blot validated the B4GalT5 protein expression in asthma model mice and with or without AAV-miR-491-5p treatment. (eh) N/Pi, WAi/Pi, WAm/Pi and WAi/WAm level was calculated. (i) Lung tissue sections were stained by HE, PAS and Masson. Scale bar, 100 μm. (jl) HE staining inflammation score, PAS glycogen staining score and Masson collagen fibrillar staining score. Data are presented as means ± SEM and three or more independent experiments were performed. Significance was calculated by one-way ANOVA followed by Tukey’s post-hoc test. *P<0.05, **P<0.01, ****P<0.0001. n = 6 mice/group.

    Abbreviations: qRT-PCR, quantitative reverse transcription polymerase chain reaction; HE, hematoxylin and eosin; PAS, periodic acid–Schiff; Pi, inner perimeter of the bronchial wall; WAi, inner area of the bronchial wall; WAm, airway smooth muscle area; N, the number of bronchial smooth muscle cells.

    Next, we examined the pathological changes in lung tissue using HE, Masson, and PAS staining. HE staining revealed increased inflammatory infiltration around the bronchi, airway wall thickening, and epithelial damage in the asthma group, while miR-491-5p-AAV intervention significantly alleviated these pathological changes. PAS staining showed a significant increase in mucin secretion and obvious goblet cell hyperplasia in the airway epithelium of the OVA group, while miR-491-5p-AAV intervention significantly suppressed excessive mucin secretion and goblet cell hyperplasia. Masson staining results confirmed increased collagen deposition and significant airway fibrosis around the airway epithelium in the asthma group, while in miR-491-5p asthma mice, collagen deposition and airway fibrosis were alleviated (Figure 6i). Furthermore, HE inflammation scores, PAS scores, and Masson collagen deposition area also confirmed that miR-491-5p significantly improved airway inflammation, excessive mucin secretion, and fibrotic pathological changes in the asthma mice (Figure 6j–l).

    ELISA results showed that compared with the control group, levels of IL-6, IL-8, and IL-1β in the BALF were significantly elevated in the OVA group, while miR-491-5p-AAV intervention markedly reduced these inflammatory cytokines (Figure 7a). In addition, miR-491-5p-AAV effectively decreased the elevated levels of OVA-specific IgE in the serum of OVA-challenged mice, suggesting that miR-491-5p can significantly alleviate OVA-induced pulmonary inflammation (Figure 7b).

    Figure 7 miR-491-5p attenuates pulmonary inflammation and oxidative stress in OVA-induced asthma mouse model (a) ELISA assay examined the IL-6, IL-8 and IL-1β level in BALF. (b) ELISA assay examined the IgE antibody level in serum. (c) Detection of ROS production in lung tissue by DCFH-DA fluorescence assay. Scale bar, 50 μm. (df) Determination of MDA, SOD and ATP in the lung tissue. Data are presented as means ± SEM and three or more independent experiments were performed. Significance was calculated by one-way ANOVA followed by Tukey’s post-hoc test. ***P<0.001, ****P<0.0001. n = 6 mice/group.

    Abbreviations: BALF, bronchoalveolar lavage fluid; IgE, immunoglobulin E; ROS, reactive oxygen species; MDA, malondialdehyde; SOD, superoxide dismutase; ATP, adenosine triphosphate.

    Finally, we measured the levels of oxidative stress and lipid peroxidation markers in the lung. The results showed that, compared to the control group, ROS fluorescence intensity and MDA levels were significantly increased in the lung tissue of the OVA group, whereas miR-491-5p-AAV intervention significantly reduced ROS fluorescence intensity and MDA levels, and quantitative analysis further confirmed this (Figure 7c and d). In addition, SOD activity in the lung tissue of the OVA group was significantly lower than that of the control group, and miR-491-5p-AAV treatment partially reversed this trend (Figure 7e). To assess the impact of miR-491-5p on mitochondrial energy metabolism, we measured ATP levels in lung tissue. Interestingly, compared to the control group, ATP levels were significantly reduced in the OVA group, and treatment with miR-491-5p-AAV partially restored ATP levels (Figure 7f).

    Discussion

    MicroRNAs function as upstream regulators in gene networks, can target multiple genes and pathways to form regulatory networks involved in the pathogenesis of various diseases, including asthma.16 This study reveals the key regulatory role of miR-491-5p in the pathogenesis of asthma and its underlying molecular mechanisms. Our findings show that miR-491-5p is significantly downregulated in the ASM of asthma patients, and its expression level is significantly negatively correlated with B4GalT5. Importantly, through a systematic in vivo and in vitro validation, we confirmed that miR-491-5p directly targets and negatively regulates B4GalT5 expression, and further elucidated the “miR-491-5p/B4GalT5” regulatory axis, which exacerbates asthma airway inflammation and remodeling through mediating oxidative stress. This core finding is further supported by clinical data: the expression of B4GalT5 is not only significantly positively correlated with the cell proliferation marker Ki67 but also with the WA%, which provides clinical evidence for the crucial role of this regulatory pathway in ASM proliferation and remodeling. This discovery offers new theoretical insights into the pathogenesis of asthma and lays an experimental foundation for the development of asthma therapeutic strategies targeting miR-491-5p.

    The pathological progression of asthma is primarily driven by chronic airway inflammation and structural remodeling, with these two processes mutually reinforcing each other in a vicious cycle. In terms of inflammation, continuous release of pro-inflammatory and chemotactic factors leads to epithelial damage, increased mucus secretion, and immune cell infiltration, further inducing airway hyperresponsiveness.17 Recurrent inflammatory stimuli trigger airway remodeling, including abnormal proliferation of ASMCs, thickening of the basement membrane, and deposition of extracellular matrix, ultimately leading to irreversible airway narrowing and progressive decline in lung function.18 Notably, mitochondrial oxidative stress serves as a key link between airway inflammation and remodeling, exacerbating this pathological process through multiple mechanisms. Specifically, mitochondrial dysfunction leads to excessive ROS production, which, on one hand, activates the NF-κB and NLRP3 inflammasome pathways to promote the cascade release of pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α, amplifying the inflammatory response;5,19 on the other hand, ROS interferes with Ca²⁺ homeostasis, inducing Ca²⁺ overload in ASMCs, which not only enhances their contractility but also significantly promotes their proliferation and phenotypic transformation.20 This vicious cycle of mitochondrial dysfunction-oxidative stress-chronic inflammation-tissue remodeling forms the core pathological basis of the persistent progression of asthma, and also provides an important pathophysiological background for the miR-491-5p/B4GalT5 regulatory axis discovered in this study.

    In this study, we identified through transcriptome sequencing that miR-491-5p is the only significantly downregulated miRNA in the airway smooth muscle tissue of asthma patients. While previous studies have reported that miR-491-5p regulates cell proliferation and differentiation in cancers such as gastric cancer and breast cancer,6,21 its role in respiratory diseases has not been explored. Notably, miR-491-5p downregulation in oxygen-glucose deprivation-treated brain microvascular endothelial cells was found to mitigate oxidative stress damage, reduce ROS levels, improve cell viability, and promote angiogenesis.22 This finding parallels our results in ASMCs. Our in vitro experiments confirmed that upregulation of miR-491-5p significantly alleviates mitochondrial damage in ASMCs, reduces ROS generation, inhibits abnormal Ca²⁺ release, and thus relieves ASMC proliferation and inflammation. To further investigate this, we used AAV-miR-491-5p for intranasal delivery to mice, and found that AAV-miR-491-5p reversed the decrease in miR-491-5p expression in the asthma model mice, significantly improving oxidative stress in the lung tissue and effectively alleviating airway inflammation and remodeling. These findings provide direct evidence for miR-491-5p as a therapeutic target for airway remodeling in asthma.

    To elucidate the intrinsic mechanism by which miR-491-5p regulates airway inflammation and remodeling in asthma, we employed bioinformatics analysis and experimental validation to identify B4GalT5 as a key target gene in miR-491-5p-mediated regulation of oxidative stress. B4GalT5, a β-1,4-galactosyltransferase, plays a pivotal role in modifying mitochondrial surface channel proteins through abnormal glycosylation, thereby triggering oxidative stress, promoting ROS production, and exacerbating mitochondrial dysfunction. Studies have shown that B4GalT5 mediates mitochondrial oxidative stress to promote myocardial hypertrophy in cardiomyocytes.12 We provide evidence supporting that in ASMCs, overexpression of B4GalT5 can reverse miR-491-5p-mediated oxidative damage, alleviating ASMC proliferation and inflammation, and supporting the therapeutic potential of miR-491-5p agonists in treating asthma airway remodeling.

    The significance of this study lies in the fact that, despite the involvement of numerous miRNAs in the pathological process of asthma, miR-491-5p exerts a unique and critical regulatory role in asthma airway smooth muscle. By integrating clinical sample analysis, molecular mechanism exploration, and animal experiments, this study not only establishes the potential of miR-491-5p as a reliable asthma biomarker but also reveals that miR-491-5p directly targets and inhibits B4GalT5 transcription, significantly reducing mitochondrial oxidative stress damage, and ultimately providing multiple protective effects on ASMCs’ abnormal proliferation, inflammation, and phenotypic transformation. These findings suggest potential clinical applications, as miR-491-5p expression levels in airway tissues may serve as a valuable diagnostic biomarker for monitoring asthma progression and preventing airway remodeling. Furthermore, the targeted delivery of miR-491-5p mimics or B4GalT5 inhibitors to airway smooth muscle cells through nanoparticle systems could offer a novel precision medicine strategy for managing refractory asthma. However, these translational prospects should be interpreted with consideration of the study’s limitations. First, the number of clinical samples used for miRNA sequencing was small (n=3), which may limit the generalizability of the results and warrants validation in larger cohorts. Second, while we identified B4GalT5 as a direct target of miR-491-5p, we did not perform direct inhibition of B4GalT5 to verify whether its knockdown would phenocopy the effects of miR-491-5p overexpression. Third, we did not explore in depth the role of mitochondrial oxidative stress in the miR-491-5p/B4GalT5-mediated regulatory axis. Further studies are required to address these important questions and to fully elucidate the underlying mechanisms.

    Conclusions

    Our study confirms that miR-491-5p regulates B4GalT5 by binding to its 3’-UTR, which alters oxidative stress levels in ASMCs, thereby leading to airway inflammation and remodeling. This study sheds light on the miR-491-5p/B4GalT5 axis as a key target in regulating airway remodeling in asthma, providing a theoretical foundation for understanding the pathogenesis of asthma and future development of innovative therapies.

    Abbreviations

    B4GalT5, β-1,4-galactosyltransfe; ASM, Airway smooth muscle; WA%, Percentage of airway wall area to total tracheal area; OVA, Ovalbumin; qRT-PCR, Quantitative real-time PCR; ROS, Reactive oxygen species; MDA, Malondialdehyde; SOD, Superoxide dismutase; ATP, Adenosine triphosphate; ASMCs, Airway smooth muscle cells; GINA, Global initiative for asthma; COPD, Chronic obstructive pulmonary disease; PBS, Phosphate-buffered saline; HRCT, High-resolution computed tomography; RIN, RNA integrity number; SPF, Specific pathogen-free; BALF, Bronchoalveolar lavage fluid; Pi, Internal perimeter; WAi, Wall area; WAm, Smooth muscle area; N, Number of smooth muscle cells; DMEM, Dulbecco’s modified eagle medium; FBS, Fetal bovine serum; WT, Wild-type; MUT, Mutant; NC, Negative control; CCK-8, Cell counting kit 8; TEM, Transmission electron microscopy.

    Data Sharing Statement

    The sequencing data generated in this study are currently under controlled access and will be made publicly available upon publication. The publicly available dataset used for correlation analysis was obtained from the Gene Expression Omnibus (GEO) database under accession number GSE119580.

    Ethic Approval

    The collection and use of human specimens in this study were approved by the Medical Ethics Committee of the People’s Hospital of Zhengzhou University (Approval No. 202407701). All procedures strictly adhered to the ethical principles outlined in the Declaration of Helsinki. Written informed consent was obtained from all participants. The privacy and confidentiality of all subjects were strictly protected throughout the study. All protocols related to animal experimental was approved by the Medical Ethics Committee of Zhengzhou University (Approval No. ZZU-LAC20240628[02]) and complied with the Helsinki Declaration and relevant guidelines for animal welfare.

    Acknowledgment

    We would like to thank Yue Liu for his suggestions when we had difficulties in conducting this study.

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Funding

    This study is supported by the Henan Province Medical Science and Technology Research and Development Plan Key Projects Jointly Constructed by the Provincial and Ministerial Departments (SBGJ202302003).

    Disclosure

    The authors declare no competing interests.

    References

    1. Porsbjerg C, Melén E, Lehtimäki L, Shaw D. Asthma. Lancet. 2023;401(10379):858–873. doi:10.1016/S0140-6736(22)02125-0

    2. Khalfaoui L, Pabelick CM. Airway smooth muscle in contractility and remodeling of asthma: potential drug target mechanisms. Expert Opin Ther Targets. 2023;27(1):19–29. doi:10.1080/14728222.2023.2177533

    3. Pokharel MD, Garcia-Flores A, Marciano D, et al. Mitochondrial network dynamics in pulmonary disease: bridging the gap between inflammation, oxidative stress, and bioenergetics. Redox Biol. 2024;70:103049. doi:10.1016/j.redox.2024.103049

    4. Ornatowski W, Lu Q, Yegambaram M, et al. Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol. 2020;36:101679. doi:10.1016/j.redox.2020.101679

    5. Albano GD, Gagliardo RP, Montalbano AM, Profita M. Overview of the mechanisms of oxidative stress: impact in inflammation of the airway diseases. Antioxidants. 2022;11(11). doi:10.3390/antiox11112237

    6. Huang W-C, Chi H-C, Tung S-L, et al. Identification of the novel tumor suppressor role of FOCAD/miR-491-5p to inhibit cancer stemness, drug resistance and metastasis via regulating RABIF/MMP signaling in triple negative breast cancer. Cells. 2021;10(10). doi:10.3390/cells10102524

    7. Ma G, Dai W, Zhang J, et al. ELK1‑mediated upregulation of lncRNA LBX2‑AS1 facilitates cell proliferation and invasion via regulating miR‑491‑5p/S100A11 axis in colorectal cancer. Int J Mol Med. 2021;48(1). doi:10.3892/ijmm.2021.4971

    8. Tong T, Zhai PS, Qin X, et al. Nuclear TOP1MT confers cisplatin resistance via pseudogene in HNSCC. J Dent Res. 2024;103(12):1238–1248. doi:10.1177/00220345241272017

    9. Liu F, Zhang H, Xie F, et al. Hsa_circ_0001361 promotes bladder cancer invasion and metastasis through miR-491-5p/MMP9 axis. Oncogene. 2020;39(8):1696–1709. doi:10.1038/s41388-019-1092-z

    10. Xing X, Yin S-Q, Li X-Q, et al. B4GALT5 inhibits CD8 + T-cell response by downregulating MHC-I level through ERAD pathway in PDAC. J Immunother Cancer. 2025;13(5):e010908. doi:10.1136/jitc-2024-010908

    11. Wu D, Sun L-Y, Chang X-Y, Zhang G-M. B4GALT5 a sialylation-related genes associated with patient prognosis and immune microenvironment in ovarian cancer and pan-cancer. J Ovarian Res. 2024;17(1):176. doi:10.1186/s13048-024-01503-3

    12. Cui S, Zhang X, Li Y, et al. UGCG modulates heart hypertrophy through B4GalT5-mediated mitochondrial oxidative stress and the ERK signaling pathway. Cell Mol Biol Lett. 2023;28(1):71. doi:10.1186/s11658-023-00484-3

    13. Global lnitiative for Asthma. Global strategy for asthma management and prevention. 2023. Available from: www.ginasthma.org. Accessed August 21, 2025.

    14. Hu L, Li L, Yan C, Cao Y, Duan X, Sun J. Baicalin inhibits airway smooth muscle cells proliferation through the RAS signaling pathway in murine asthmatic airway remodeling model. Oxid Med Cell Longev. 2023;2023:4144138. doi:10.1155/2023/4144138

    15. Wang Q, Ji J, Xiao S, Wang J, Yan X, Fang L. Explore alteration of lung and gut microbiota in a murine model of OVA-induced asthma treated by CpG oligodeoxynucleotides. J Inflamm Res. 2025;18:445–461. doi:10.2147/JIR.S487916

    16. Liu S, Chen X, Zhang S, et al. miR‑106b‑5p targeting SIX1 inhibits TGF‑β1‑induced pulmonary fibrosis and epithelial‑mesenchymal transition in asthma through regulation of E2F1. Int J Mol Med. 2021;47(3). doi:10.3892/ijmm.2021.4857

    17. Ford ML, Reza MI, Ruwanpathirana A, Sathish V, Britt RD. Integrative roles of pro-inflammatory cytokines on airway smooth muscle structure and function in asthma. Immunol Rev. 2025;330(1):e70007. doi:10.1111/imr.70007

    18. Varricchi G, Brightling CE, Grainge C, Lambrecht BN, Chanez P. Airway remodelling in asthma and the epithelium: on the edge of a new era. Europ resp J. 2024;63(4):2301619. doi:10.1183/13993003.01619-2023

    19. Chen A, Huang H, Fang S, Hang Q. ROS: a “booster” for chronic inflammation and tumor metastasis. Biochim Biophys Acta Rev Cancer. 2024;1879(6):189175. doi:10.1016/j.bbcan.2024.189175

    20. Mahapatra C, Thakkar R, Kumar R. Modulatory impact of oxidative stress on action potentials in pathophysiological states: a comprehensive review. Antioxidants. 2024;13(10):1172. doi:10.3390/antiox13101172

    21. Kang W, Zhang J, Huang T, et al. NOTCH3, a crucial target of miR-491-5p/miR-875-5p, promotes gastric carcinogenesis by upregulating PHLDB2 expression and activating Akt pathway. Oncogene. 2021;40(9):1578–1594. doi:10.1038/s41388-020-01579-3

    22. Tang W, Guo Z-D, Chai W-N, et al. Downregulation of miR-491-5p promotes neovascularization after traumatic brain injury. Neural Regen Res. 2022;17(3):577–586. doi:10.4103/1673-5374.314326

    Continue Reading

  • ‘Want to go one better’ – Chirag Shetty, Satwiksairaj Rankireddy set sights on making final at BWF World Championships 2025 – Olympics.com

    ‘Want to go one better’ – Chirag Shetty, Satwiksairaj Rankireddy set sights on making final at BWF World Championships 2025 – Olympics.com

    1. ‘Want to go one better’ – Chirag Shetty, Satwiksairaj Rankireddy set sights on making final at BWF World Championships 2025  Olympics.com
    2. Sindhu, Lakshya dip tempers Indian hopes  Hindustan Times
    3. Badminton World Championships: Where to watch livestream for India matches  India Today
    4. Sen, Sindhu face stern test at BWF Worlds  Press Trust of India
    5. Lakshya Sen vs Shi Yuqi World Championships 2025: Check live streaming and head-to-head details  India TV News

    Continue Reading

  • Four journalists among 20 dead in Israeli strike on hospital, Gaza officials say

    Four journalists among 20 dead in Israeli strike on hospital, Gaza officials say

    Who were the journalists?published at 12:29 British Summer Time

    Reuters news agency has said its cameraman, Hussam al-Masri, was among those killed.

    It adds that one of their contractors was also injured in the strikes.

    Cameraman al-Masri was killed in the first strike, Reuters reports, and photographer Hatem Khaled was wounded in a second strike.

    “We are devastated to learn of the death of Reuters contractor Hussam al-Masri and injuries to another of our contractors, Hatem Khaled, in Israeli strikes on the Nasser hospital in Gaza today,” a Reuters spokesperson said in a statement.

    The Associated Press (AP) says Mariam Dagga, a freelance journalist working for it, was also killed. AP said it was “shocked and saddened” by the 33-year-old’s death.

    The others are said to have been Mohammed Salama, working for Al Jazeera, and photographer Moaz Abu Taha.

    US TV network NBC said Taha did not work for it, as had been initially reported.

    Continue Reading

  • Mayo Clinic ramps up spinout creation as funding cuts bite –

    Mayo Clinic ramps up spinout creation as funding cuts bite –

    The Mayo Clinic has hired entrepreneurs to speed the commercialisation of its medical research as funding from the government declines.

    Audrey Greenberg, one of the Mayo Clinic’s new venture partners

    US university hospital system Mayo Clinic, known for its pioneering healthcare research, is homing in on creating and funding companies based on its own healthcare IP, as government funding for health research wanes in the US under the Trump administration.

    “Mayo is at a time of tight capital markets and increased scrutiny from an [National Institutes of Health] funding perspective and a flight to quality in the capital markets. So, Mayo is really doubling down and standing behind its innovative technology,” says Audrey Greenberg, a venture partner hired under new programme aimed at boosting venture building.

    The Mayo Clinic launched the programme in May to bring in external entrepreneurs and experienced investors who could build companies based intellectual property from the Mayo Clinic. The initiative builds on the role of Mayo Clinic Ventures, the commercialisation arm of the Mayo Clinic which licenses and creates ventures based on the medical centre’s IP.

    The extra resources that Mayo is putting into building ventures comes amid budget cuts at the National Institutes of Health (NIH), the medical research agency of the US government, which provides grants for medical research. The US administration has reportedly terminated nearly $10bn in research grants since president Donald Trump took office at the start of 2025.

    Reduced government funding, combined with a decrease in venture capital funding for healthcare, is spurring US university hospital systems like the Mayo Clinic to focus on their own commercislisation efforts.

    Greenberg is an experienced entrepreneur, having founded and scaled healthcare companies. She also founded the Center for Breakthrough Medicines, a manufacturer of cell and gene therapies. Other newly hired entrepreneurs are Amy DuRoss, co-founder and CEO of Vineti, a developer of advanced therapy supply chain software, and Brian Poger, founding CEO of Alight Health Exchange and Benefitter, a benefits platform for small businesses.

    The venture partners team will focus on building companies particularly in the fields of diagnostics, AI digital health and tech-enabled therapeutics where it is early enough to establish sustainable companies and platforms that have access to patients. 

    “Ninety percent of our time will be spent on venture creation based on Mayo innovations,” Greenberg says. “But if there’s a compelling strategic opportunity to invest outside of Mayo, we’re prepared to deploy capital there as well.” 

    One element that drew Greenberg to the venture partners programme was the Mayo Clinic’s focus on patient healthcare. She says she wants to prevent innovative ideas falling to the wayside. “I want to commercialise innovations that improve outcomes, reduce costs, or increase system efficiency, and make sure they don’t fall through the cracks.” 

    The programme is focusing on innovations such as AI-aided predictive diagnostics which may allow diseases to be treated in new or more effective ways, as well as in personalised tools and integrated digital platforms with workflow automation. 

    Another bucket of innovation is AI-driven clinical decision support. “These tools provide instant, continuous feedback loops from equipment, data and processes to better predict outcomes and improve decision-making,” she says.  

    The venture partners are also open to co-investing with CVCs. “We welcome partnerships with corporate investors, especially those with strategic alignment to help catalyse growth and amplify impact,” Greenberg says.


     Mayo Clinic is an active investor in startups through its Mayo Clinic Ventures arm. See all startups that the company has backed in the CVC Funding Round Database

    Mayo Clinic Ventures deals 2025

    Continue Reading

  • Dominican Republic on to eighthfinals after 3-0 over Mexico

    It was Mexico’s 25 unforced errors (against 14 from Dominican Republic) that helped their opponents the most during the match and even the game winning point that came on a mistaken serve exemplified that. The Caribbean Queens also outplayed their rivals in attack with 42 successful swings against 32, and in blocking with six kill blocks against three. Outside hitter and captain Brayelin Martinez authored three of those kill blocks, fired an ace and spiked at a 69% success rate to contribute a match-high 22 points to the Dominican win. Her cross-court teammate Yonkaira Pena added another 11 points, all in swings.

    “We are very happy about qualifying. To make it to the second round was an important goal. Now we have to prepare for the next goal, which is to finish first in the pool,” Dominican Republic’s Brazilian head coach Marcos Kwiek told VBTV. “We prepared very well for this game. The third set was the hardest we had, but we know Mexico, we’ve been playing them for a while, so we managed to neutralize them and we are just happy we did a good job.”

    The third set was the only one, in which Mexico maintained a lead in the score for a while. Serving was the only scoring element, in which the North Americans slightly outplayed their NORCECA rivals in this match, by 3-2 in aces. Opposite Sofia Maldonado was their best scorer of the game with 13 points, all in attack.

    Continue Reading

  • Dawn of War IV’ release date window, gameplay details, more

    Dawn of War IV’ release date window, gameplay details, more

    One of the biggest reveals during Opening Night Live at Gamescom 2025 was the return of Dawn of War, with Deep Silver specifically announcing Warhammer 40,000: Dawn of War IV. The series is heading back to its roots after almost a decade and is set to feature a ton of missions to tackle.

    Here is a full breakdown of what we know:

    ‘Warhammer 40,000: Dawn of War IV’ release date window

    Dawn of War IV is set to be released in 2026 on PC via Steam. There are no details about a console version yet, but a release after the PC version is likely.

    ‘Warhammer 40,000 Dawn of War IV’ gameplay details

    Dawn of War IV sees the series return to its roots after some big changes with Dawn of War III back in 2017. This entry features classic base-building, large armies, and deep strategic gameplay all set on the planet Kronus, approximately 200 years after the events of Dawn of War: Dark Crusade.

    Space Marines, Orks, Necrons, and the Adeptus Mechanicus are all playable totalling more than 70 missions in total with different sides of the story to be discovered depending on which faction you play as.

    There are more than 10 playable Commanders, more than 110 units and buildings, many of which are customizable, as well as an in-game editor for player-created content and a Painter tool for unit customization. On top of that, Skirmish, co-op, competitive multiplayer, and Last Stand are all here, allowing you to take on others or work with them to survive.

    We will have even more on the game as we get closer to launch, so be sure to stay tuned. For even more on Gamescom, check out our roundup of all the announcements during Opening Night Live.


    Continue Reading

  • Is AI Use Causing Endoscopists to Lose Their Skills?

    Is AI Use Causing Endoscopists to Lose Their Skills?

    Routine use of artificial intelligence (AI) may lead to a loss of skills among clinicians who perform colonoscopies, thereby affecting patient outcomes, a large observational study suggested.

    “The extent and consistency of the adenoma detection rate (ADR) drop after long-term AI use were not expected,” study authors Krzysztof Budzyń, MD, and Marcin Romańczyk, MD, of the Academy of Silesia, Katowice, Poland, told Medscape Medical News. “We thought there might be a small effect, but the 6% absolute decrease — observed in several centers and among most endoscopists — points to a genuine change in behavior. This was especially notable because all participants were very experienced, with more than 2000 colonoscopies each.”

    Another unexpected result, they said, “was that the decrease was stronger in centers with higher starting ADRs and in certain patient groups, such as women under 60. We had assumed experienced clinicians would be less affected, but our results show that even highly skilled practitioners can be influenced.”

    The study was published online in The Lancet Gastroenterology & Hepatology.

    ADR Reduced After AI Use

    To assess how endoscopists who used AI regularly performed colonoscopy when AI was not in use, researchers conducted a retrospective, observational study at four endoscopy centers in Poland taking part in the ACCEPT trial.

    These centers introduced AI tools for polyp detection at the end of 2021, after which colonoscopies were randomly assigned to be done with or without AI assistance.

    The researchers assessed colonoscopy quality by comparing two different phases: 3 months before and 3 months after AI implementation. All diagnostic colonoscopies were included, except for those involving intensive anticoagulant use, pregnancy, or a history of colorectal resection or inflammatory bowel disease.

    The primary outcome was the change in the ADR of standard, non-AI-assisted colonoscopy before and after AI exposure.

    Between September 2021 and March 2022, a total of 2177 colonoscopies were conducted, including 1443 without AI use and 734 with AI. The current analysis focused on the 795 patients who underwent non-AI-assisted colonoscopy before the introduction of AI and the 648 who underwent non-AI-assisted colonoscopy after.

    Participants’ median age was 61 years, and 59% were women. The colonoscopies were performed by 19 experienced endoscopists who had conducted over 2000 colonoscopies each.

    The ADR of standard colonoscopy decreased significantly from 28.4% (226 of 795) before the introduction of AI to 22.4% (145 of 648) after, corresponding to a 20% relative and 6% absolute reduction in the ADR.

    The ADR for AI-assisted colonoscopies was 25.3% (186 of 734).

    The number of adenomas per colonoscopy (APC) in patients with at least one adenoma detected did not change significantly between the groups before and after AI exposure, with a mean of 1.91 before vs 1.92 after. Similarly, the number of mean advanced APC was comparable between the two periods (0.062 vs 0.063).

    The mean advanced APC detection on standard colonoscopy in patients with at least one adenoma detected was 0.22 before AI exposure and 0.28 after AI exposure.

    Colorectal cancers were detected in 6 (0.8%) of 795 colonoscopies before AI exposure and in 8 (1.2%) of 648 after AI exposure.

    In multivariable logistic regression analysis, exposure to AI (odds ratio [OR], 0.69), patient’s male sex (OR, 1.78), and patient age at least 60 years (OR, 3.60) were independent factors significantly associated with ADR.

    In all centers, the ADR for standard, non-AI-assisted colonoscopy was reduced after AI exposure, although the magnitude of ADR reduction varied greatly between centers, according to the authors.

    “Clinicians should be aware that while AI can boost detection rates, prolonged reliance may subtly affect their performance when the technology is not available,” Budzyń and Romańczyk said. “This does not mean AI should be avoided — rather, it highlights the need for conscious engagement with the task, even when AI is assisting. Monitoring one’s own detection rates in both AI-assisted and non-AI-assisted procedures can help identify changes early.”

    “Endoscopists should view AI as a collaborative partner, not a replacement for their vigilance and judgment,” they concluded. “Integrating AI effectively means using it to complement, not substitute, core observational and diagnostic skills. In short, enjoy the benefits of AI, but keep your skills sharp — your patients depend on both.”

    Omer Ahmed, MD, of University College London, London, England, gives a similar message in a related editorial. The study “compels us to carefully consider the effect of AI integration into routine endoscopic practice,” he wrote. “Although AI continues to offer great promise to enhance clinical outcomes, we must also safeguard against the quiet erosion of fundamental skills required for high-quality endoscopy.”

    ‘Certainly a Signal’

    Commenting on the study for Medscape Medical News, Rajiv Bhuta, MD, assistant professor of clinical gastroenterology and hepatology at Temple University and a gastroenterologist at Temple University Hospital, both in Philadelphia, said, “On the face of it, these findings would seem to correlate with all our lived experiences as humans. Any skill or task that we give to a machine will inherently ‘de-skill’ or weaken our ability to perform it.”

    “The only way to miss a polyp is either due to lack of attention/recognition of a polyp in the field of view or a lack of fold exposure and cleansing,” said Bhuta, who was not involved in the study. “For AI to specifically de-skill polyp detection, it would mean the AI is conditioning physicians to pay less active attention during the procedure, similar to the way a driver may pay less attention in a car that has self-driving capabilities.”

    That said, he noted that this is a small retrospective observational study with a short timeframe and an average of fewer than 100 colonoscopies per physician.

    “My own ADR may vary by 8% or more by random chance in such a small dataset,” he said. “It’s hard to draw any real conclusions, but it is certainly a signal.”

    The issue of de-skilling goes beyond gastroenterology and medicine, he noted. “We have invented millions of machines that have ‘de-skilled’ us in thousands of small ways, and mostly, we have benefited as a society. However, we’ve never had a machine that can de-skill our attention, our creativity, and our reason.”

    “The question is not whether AI will de-skill us but when, where, and how do we set the boundaries of what we want a machine to do for us,” he said. “What is lost and what is gained by AI taking over these roles, and is that an acceptable trade-off?”

    The study was funded by the European Commission and the Japan Society for the Promotion of Science. Budzyń, Romańczyk, and Bhuta declared having no competing interests. Ahmed declared receiving medical consultancy fees from Olympus, Odin Vision, Medtronic, and Norgine.

    Marilynn Larkin, MA, is an award-winning medical writer and editor whose work has appeared in numerous publications, including Medscape Medical News and its sister publication MDedge, The Lancet (where she was a contributing editor), and Reuters Health.

    Continue Reading