Blog

  • Close approach of asteroid 2025 FA22

    Close approach of asteroid 2025 FA22

    Continue Reading

  • The new economics of mobile ad personalization

    The new economics of mobile ad personalization

    App publishers face a fundamental contradiction, with users increasingly wanting personalized experiences while simultaneously distrusting the data collection that makes personalization possible.

    Data from a recent strategy session featuring Verve’s SVP & GM of Marketplace Aviran Edery alongside industry experts from Singular, ID5, and GeoEdge, plus Verve’s new app privacy report based on 4,000 mobile users, reveals how this tension is reshaping the economics of mobile advertising, forcing publishers to rethink everything from consent timing to targeting strategies.

    Three-quarters of consumers now prefer watching ads over paying for content, up from two-thirds last year. Yet 65% express growing concern about their data being used to train AI systems. This paradox creates both opportunity and risk for publishers who’ve built monetization strategies around data-driven personalization.

    The geography of trust

    Privacy attitudes aren’t uniform across markets, creating new strategic considerations for global publishers. UK users have warmed to data sharing by three percentage points year-over-year, while US users show a sharp five-point decline in comfort levels. This divergence reflects different regulatory environments and cultural attitudes toward privacy, suggesting one-size-fits-all approaches may be leaving money on the table.

    Publishers operating across both markets face a complex optimization problem — customize privacy strategies by region or maintain operational simplicity with potentially suboptimal results. The data suggests customization may be worth the investment, particularly as regulatory frameworks continue to diverge globally.

    Redefining personalization

    The industry’s approach to personalization has reached a breaking point. Traditional hyper-targeting, where users see ads for products they’ve already researched across multiple touchpoints, has devolved into what users perceive as surveillance. The economics no longer favor this approach when churn costs are factored against incremental CPM gains.

    A more sustainable model focuses on contextual relevance over invasive tracking. Instead of knowing a user searched for specific red socks and following them across the internet, effective personalization recognizes they’re interested in fashion accessories within a shopping app context. This “gentle personalization” approach delivers relevance without crossing into creepy territory.

    Gaming apps provide a clear example: showing similar games to users already engaged with mobile gaming makes contextual sense and feels natural. Showing laptop ads to someone playing a puzzle game during their commute does not. The distinction seems obvious, yet measurement data shows many publishers still optimize for data collection over user experience.

    Music streaming services like Spotify demonstrate how personalization can enhance rather than exploit user relationships. Discover Weekly playlists feel like curation rather than surveillance because they’re built on listening behavior within the platform and delivered as value-added features, not advertising.

    The timing arbitrage

    Most publishers request permissions at the worst possible moment, namely immediately after app installation, before demonstrating any value. This approach optimizes for compliance rather than conversion, treating consent as a hurdle to clear rather than a relationship to build.

    Smart publishers are discovering a timing arbitrage opportunity. By delaying permission requests until after users experience app value, they can dramatically improve consent rates while building stronger relationships. This progressive consent model starts with basic functionality and contextual advertising, then requests additional permissions as users become more engaged.

    The strategy requires patience but pays dividends in user lifetime value. A user who grants permission on day ten after experiencing app benefits is far more likely to remain opted-in than someone who consents on day one out of confusion or resignation.

    Ad quality as revenue protection

    Publishers often treat ad quality as a compliance checkbox rather than a revenue protection mechanism. This mindset misses the crucial economic reality that user experience and ad experience are indistinguishable from the user’s perspective. A single bad ad impression can destroy months of carefully built trust and lifetime value.

    The most successful publishers are shifting from “set it and forget it” ad operations to active quality monitoring. They recognize that users blame the app, not the ad network, when they encounter malicious or misleading advertisements. This responsibility can’t be outsourced to demand partners who may have different quality standards or economic incentives.

    Proactive ad quality management requires operational investment but protects against catastrophic user churn. Publishers report that implementing systematic ad monitoring and filtering sees immediate improvements in user retention and app store ratings, often offsetting any short-term revenue impact from filtering out problematic demand.

    The AI training disclosure imperative

    The 65% concern rate around AI training represents a new frontier in user privacy expectations. Unlike advertising personalization, which users can rationalize as value exchange, AI training feels extractive without clear benefit. Publishers who address this concern proactively gain competitive advantage over those who ignore it.

    The solution involves separating AI training consent from advertising personalization in user interfaces. Users should be able to opt into personalized ads while opting out of AI training, or vice versa. This granular control acknowledges that different data uses carry different risk-benefit calculations for users.

    Early adopters of transparent AI training disclosure report higher overall trust scores and better retention metrics. The approach requires additional development work but positions publishers ahead of likely regulatory requirements while building user goodwill.

    Implementation roadmap

    Publishers ready to optimize their personalization economics should start with three immediate actions. First, audit current consent timing and test delayed permission requests against existing day-one approaches. Second, implement systematic ad quality monitoring rather than relying on partner assurances. Third, separate AI training consent from advertising personalization in user interfaces.

    Medium-term initiatives should include developing region-specific privacy approaches for global publishers and optimizing contextual targeting capabilities. The goal is delivering relevant ads based on immediate context rather than extensive behavioral tracking.

    Long-term success requires building measurement frameworks that track trust metrics alongside traditional monetization KPIs. Publishers need visibility into how privacy decisions impact user lifetime value, not just immediate CPMs.

    The bottom line

    The economics of mobile ad personalization are shifting from data extraction to value creation. Publishers who recognize this transition early will build sustainable competitive advantages over those clinging to surveillance-based models.

    The opportunity is significant because most publishers haven’t made this transition yet. Steady 15% opt-out rates across the industry suggest users aren’t abandoning personalized advertising entirely, they’re just demanding better value exchange and transparent practices.

    The winners in this new landscape will be publishers who treat privacy as a product feature rather than a regulatory burden, who optimize for long-term user relationships rather than short-term data collection, and who recognize that sustainable monetization requires sustainable trust.

    Catch the full strategy session here.

    Continue Reading

  • Association Between Body Roundness Index and Chronic Obstructive Pulmo

    Association Between Body Roundness Index and Chronic Obstructive Pulmo

    Introduction

    Chronic obstructive pulmonary disease (COPD) is a group of progressive respiratory disorders caused by chronic airway inflammation, and it is characterized by airway obstruction or persistent airflow limitation.1 Major symptoms of this disease include chronic cough, sputum production, and dyspnea. Clinically, COPD often exhibits recurrent exacerbations and is difficult to cure, and the common diseases of COPD include asthma, emphysema, and chronic bronchitis.2 Progressive decline in lung function leads to reduced physical activity (PA) in individuals with COPD and multiple complications, including congestive heart failure (CHF) and diabetes, as well as psychological issues (such as severe anxiety and depression). These factors severely impair patients’ quality of life and can be life-threatening. The World Health Organization reports that nearly 299 million individuals worldwide suffer from COPD. It stands as the fourth major cause of death worldwide,3 with incidence rates rising every year. For example, in 2017, the prevalence of COPD in the United States was 15.2% among current cigarette smokers, 7.6% among former smokers, and 2.8% among adults who had never smoked.4 COPD typically develops insidiously and is often overlooked during the early stages. Patients usually receive medical attention only after symptoms become pronounced, thereby resulting in a poor prognosis.5,6 COPD also requires substantial healthcare resources, imposing a considerable burden on global medical and public health systems. Therefore, identifying factors influencing COPD and developing effective prevention strategies to reduce its incidence is of great importance in alleviating this global health and socioeconomic burden.

    Obesity, a prevalent metabolic disorder, is linked to a range of systemic complications. It can cause functional impairments in organs and tissues, ultimately resulting in organic lesions. The relationship between obesity and COPD has received growing attention.7 In particular, the accumulation of visceral fat (VF) in individuals with obesity may elevate the risk of COPD.8 Obesity may trigger pulmonary disease by inducing oxidative stress (OS) and systemic inflammation.7,9 Thus, a reliable indicator for assessing VF is essential for effective chronic disease control and enhancing quality of life.

    The Body Roundness Index (BRI), a novel anthropometric metric based on waist circumference (WC) and height, can comprehensively reflect visceral adiposity and body fat percentage.10 Increasing evidence demonstrates that BRI holds great potential for disease risk stratification. Elevated BRI levels are associated with many chronic diseases, such as chronic kidney disease (CKD), diabetes, and cardiovascular diseases (CVD), including atherosclerosis and hypertension (HP).11,12 Nonetheless, the relationship between BRI and COPD remains unclear. Using data from the National Health and Nutrition Examination Survey (NHANES) 2013–2018, this study aims to examine the association between BRI and COPD.

    Materials and Methods

    Study Design and Population

    Conducted by the National Center for Health Statistics (NCHS), the NHANES employs a stratified, multistage sampling design and various data collection methods to analyze the nutritional and health status of American children and adults. All NHANES protocols were approved by the Ethics Review Board of NCHS. Written informed consent was provided by all enrolled individuals. To ensure participant confidentiality, all data were de-identified. According to the Ethical Review Methods for Life Science and Medical Research Involving Human Participants, Article 32 exempts certain research from requiring ethical approval under specific conditions. Research utilizing legally obtained public data or data derived from non-intrusive observation of public behavior does not require ethical approval.

    Data from three consecutive NHANES cycles (2013–2018) were collected. A total of 29,400 individuals were initially included. Individuals were excluded if they were under 20 years of age (n = 12,343), had incomplete BRI data (n = 1766), or lacked information on key covariates, including smoking status (n = 9), alcohol consumption (n = 1011), educational background (n = 9), marital status (n = 3), and PA (n = 5). Ultimately, 14,254 individuals were enrolled (Figure 1).

    Figure 1 Flowchart of participant selection.

    COPD Outcomes

    COPD was defined according to affirmative responses to any of the following self-reported questions: “Have you been diagnosed with emphysema?”, “Have you been diagnosed with chronic bronchitis?”, or “Has a doctor ever informed you that you have COPD?” Individuals who answered “no” to all three questions were considered non-COPD.

    Calculation of Anthropometric Indices

    BRI was treated as the independent variable. According to the BRI calculation formula proposed by Tomas et al,10 data on WC and height were extracted from the anthropometric measurements in NHANES for the calculation.


    Assessment of Covariates

    A wide range of lifestyle, demographic, and health-related variables were collected: alcohol consumption, poverty income ratio (PIR), sex, total cholesterol, educational background, total energy intake (TEI), race/ethnicity, high-density lipoprotein cholesterol (HDL), protein, total sugar, age, HP, marital status, height, carbohydrate, dietary fiber (DF), CVD, total fat, smoking status, vigorous and moderate recreational activities, weight, eosinophil count, diabetes, white blood cell count, monocyte count, WC, and BMI.

    Smoking status was categorized as never smoked, current smoker, or former smoker based on whether the individuals had smoked more than 100 cigarettes over a lifetime and their duration since quitting. Individuals were defined as having diabetes according to the following criteria: self-reported physician diagnosis of diabetes, fasting blood glucose >126 mg/dL, administration of glucose-lowering medication or insulin, or glycated hemoglobin (HbA1c) ≥6.5%.13 HP was defined according to the following criteria: self-reported HP, current use of antihypertensive medication, average systolic blood pressure ≥130 mmHg or average diastolic blood pressure ≥80 mmHg across three measurements.14 CVD was defined if individuals reported the following physician diagnoses: coronary heart disease, CHF, heart attack, or stroke.13

    Statistical Analysis

    Following NHANES methodological guidelines, this study applied the recommended multistage probability sampling weights to account for the complex survey design during statistical analyses. BRI was divided into four quartiles based on its distribution: Q1 (1.167–3.965), Q2 (3.965–5.282), Q3 (5.282–6.940), and Q4 (6.940–23.482). The Kolmogorov–Smirnov test was used to assess the normality of continuous variables. Given that these continuous variables were non-normally distributed, they were expressed as medians and interquartile ranges, and between-group comparisons were conducted using the Kruskal–Wallis test. Categorical variables were expressed as frequencies and percentages, and between-group comparisons were made using weighted Chi-square tests. Multicollinearity among covariates was examined, and variables with a variance inflation factor (VIF) ≥ 5 were excluded. Weighted logistic regression (WLR) models were employed to investigate the association between BRI and COPD prevalence. Three models were constructed: Model 1 was unadjusted; Model 2 was adjusted for race, sex, and age; Model 3 was further adjusted for educational background, marital status, PIR, TEI, DF, smoking status, alcohol consumption, moderate PA, diabetes, HP, CVD,15 direct HDL cholesterol, and eosinophil percentage. Restricted cubic spline (RCS) analysis with five knots was employed to investigate the dose-response relationship between BRI and COPD prevalence. Subgroup analyses stratified by sex, smoking status (never, current, former), alcohol consumption, diabetes, HP, and CVD were conducted to test the robustness of the findings. Statistical significance was defined as two-sided p-values less than 0.05. R version 4.4.2 was used to conduct statistical analysis.

    Result

    Baseline Characteristics of Study Population

    Table 1 illustrates the basic characteristics of the included 14,254 individuals (stratified by BRI quartiles). The overall prevalence of COPD was 8.3%, corresponding to a weighted estimate of 210,015,248 individuals. The median age of individuals was 48 years, with 49% men and 51% women. BRI was divided into four quartiles: Q1 (1.167–3.965), Q2 (3.965–5.282), Q3 (5.282–6.940), and Q4 (6.940–23.482).

    Table 1 Baseline Characteristics of Participants

    The highest BRI quartile had a greater proportion of women and Mexican Americans in comparison to the lowest BRI quartile. Moreover, higher BRI levels were associated with lower engagement in vigorous PA, older age, and being widowed. Moreover, the highest BRI group exhibited a greater prevalence of CVD and diabetes.

    Associations Between BRI and the Likelihood of COPD

    The association between BRI and COPD prevalence was examined using WLR models (Table 2). For continuous BRI, the results indicated that BRI was positively associated with COPD prevalence. In Model 1 (unadjusted), each unit increase in BRI was associated with a 16.8% increase in the likelihood of COPD (P < 0.001). In model 2 (adjusted for race/ethnicity, sex, and age), this association remained stable (OR = 1.141, 95% CI: 1.106–1.176, P < 0.001). In model 3 (further adjusted for comorbidities, dietary intake, and laboratory indicators), the association persisted, although slightly attenuated (OR = 1.085; 95% CI: 1.036–1.136; P = 0.002).

    Table 2 WLR Analysis of BRI and COPD

    For categorical BRI, Model 1 demonstrated that individuals in the highest BRI quartile had a 2.972-fold higher likelihood of COPD in comparison to those in the lowest quartile (P < 0.001). This association remained significant in Model 2 (OR = 2.162; 95% CI: 1.728–2.706; P < 0.001) and Model 3 (OR = 1.466; 95% CI: 1.091–1.969; P = 0.015).

    RCS Analysis

    RCS analysis examined the dose-response relationship between BRI and the likelihood of developing COPD. The results revealed an approximately linear positive association between BRI and the likelihood of developing COPD (P for nonlinearity = 0.155) (Figure 2).

    Figure 2 The association between BRI and the likelihood of developing COPD through RCS analysis. The RCS curve was adjusted for race/ethnicity, sex, PIR, educational background, marital status, age, TEI, DF, smoking status, alcohol consumption, diabetes, HP, moderate PA, direct HDL cholesterol, and eosinophil percentage.

    Subgroup Analysis

    The robustness of the association between BRI and COPD prevalence was assessed by subgroup analyses based on sex, smoking status, alcohol consumption, age, diabetes, HP, and CVD. The results demonstrated a significant association between BRI and the likelihood of developing COPD in all subgroups except for former smokers and individuals with CVD. In addition, interaction tests revealed substantial differences in COPD prevalence across subgroups stratified by sex, HP, and CVD (Figure 3).

    Figure 3 The association between BRI and COPD prevalence through subgroup analyses.

    Discussion

    This cross-sectional study, which involved 14,254 individuals, suggested that higher BRI levels were positively associated with COPD prevalence in the US adult population. This association was confirmed to be linear based on RCS analysis. Although no threshold effect was detected in the RCS analysis, it was observed that when BRI exceeded 5.332, COPD prevalence increased significantly with rising BRI. Subgroup analyses further supported the robustness of these findings. Interaction tests indicated that sex, HP, and CVD significantly modified this association. These results suggest that maintaining an appropriate BRI level is particularly important for COPD prevention. Moreover, BRI, due to its cost-effectiveness and efficiency, may hold substantial value in large-scale COPD screening and risk stratification.

    Obesity, as a major contributor to the risk of COPD, has garnered increasing research interest. Prior studies mainly employed BMI to quantify obesity, but BMI cannot differentiate between fat mass (FM) and fat-free mass (FFM). Since FFM and FM exert distinct effects on pulmonary physiology, Zhang et al highlighted the necessity of investigating the association between fat distribution and COPD risk.16 Although WC reflects abdominal fat accumulation (AFA), it does not distinguish between subcutaneous and visceral fat. The A Body Shape Index may outperform BMI and WC in predicting ACM, but is less reliable for predicting chronic diseases.17–19 In contrast, BRI, which integrates WC and height into a cylindrical model, more accurately estimates abdominal obesity.20–22 Current studies have demonstrated relationships between BRI and obstructive respiratory diseases. BRI is a superior predictor of obstructive sleep apnea.23 Xu et al demonstrated that both weight-adjusted waist index (WWI) and BRI are independent factors influencing asthma risk, with BRI exhibiting better predictive performance than WWI.24 These conclusions support the utility of BRI in early COPD detection and risk stratification. Therefore, timely identification and intervention in individuals with elevated BRI may help slow disease progression and improve their quality of life.

    Several mechanisms may underlie the association between BRI and COPD. Individuals with high BRI tend to have significant AFA, which increases the COPD risk. This finding has been supported by previous research.25,26 Lam et al reported that central obesity is associated with both obstructive and restrictive ventilatory impairments in COPD.26 This may be attributed to obesity-induced systemic inflammation, which can impair the normal structure and function of lung tissue.27 Abnormal accumulation of visceral adipose tissue (VAT) can cause adipocyte hypoxia by impairing ventilation and reducing tissue oxygenation,7,15 ultimately triggering inflammatory responses. The activation of immune cells (such as neutrophils, macrophages, and eosinophils) contributes to pulmonary inflammation.28,29 Furthermore, VAT is recognized as an active endocrine organ.28 In individuals with obesity, VAT homeostasis is disrupted, thereby leading to abnormal secretion of adipokines. This includes elevated levels of pro-inflammatory mediators such as leptin, IL-6, and TNF-α, and decreased levels of anti-inflammatory factors such as adiponectin. These imbalances can further stimulate the NOD-like receptor family pyrin domain-containing 3 inflammasome and IL-1β signaling,28,30,31 potentially accelerating airway remodeling, impairing lung function, and ultimately contributing to COPD progression.

    Furthermore, elevated levels of chronic low-grade inflammatory mediators related to obesity, such as TNF-α, leptin, and IL-6 secreted by adipose tissue, are associated with neutrophil-mediated OS responses.9 Neutrophil activation contributes to the depletion of systemic antioxidants by releasing reactive oxygen species (ROS),32 thereby reducing pulmonary defense capacity. Moreover, it can exacerbate damage to the alveolar wall by releasing proteases. In addition, ROS can activate inflammation-related signaling pathways,32,33 alter the extracellular matrix, and stimulate goblet cell activation, thereby resulting in mucus hypersecretion and subsequent airway obstruction. Current evidence demonstrates that neutrophil elastases damage elastic fibers and mucociliary structures in the lungs, ultimately impairing mucus clearance and promoting goblet cell metaplasia and mucin production.9 These effects further reduce pulmonary compliance and increase airway resistance, thereby elevating COPD risk.

    Moreover, inadequate PA also contributes to COPD progression among individuals with obesity. In individuals with COPD, obesity may further impair ventilation through such mechanical constraints as limited diaphragmatic movement and increased chest wall resistance.7,34,35 Additionally, these constraints can elevate the risk of comorbid conditions, including metabolic syndrome and CVD, thus indirectly accelerating disease deterioration.15,30

    According to subgroup analyses, no significant association between BRI and COPD was observed in participants with CVD or in former smokers, suggesting that the generalizability of BRI may vary across populations. The lack of a significant association in the CVD subgroup may be attributable to residual confounding from unmeasured factors, such as medication interventions or complications, or to potential reverse causality.36 After smoking cessation, systemic inflammation persisted in patients with COPD, whereas it declined in healthy former smokers.37 This difference may affect the BRI–COPD relationship through pathways involving fat and lung function. Existing research reveals that although smoking cessation is frequently accompanied by weight gain, it does not lead to an elevated risk of COPD.38 These findings warrant further investigation in prospective cohort studies. In addition, sex, HP, and CVD significantly modified the association between BRI and the odds of having COPD. The association between obesity and CVD may be mediated by shared intermediate metabolic risk factors, including impaired glucose tolerance, insulin resistance, HP, and hypertriglyceridemia, all of which contribute to adverse cardiovascular events.39,40 In the CVD subgroup, several plasma markers are linked to obesity.41 For example, abnormally low natriuretic peptide levels are associated with an increase in total white adipose tissue mass.42 These factors may partly disrupt the BRI–COPD relationship, distinguishing this subgroup from individuals without CVD. A U.S.-based study suggests that men are more prone to exhibit AFA.43 Abdominal fat, particularly VAT, increases COPD risk through metabolic and inflammatory mechanisms.44

    This study has several strengths. First, the generalizability of our results was enhanced by incorporating a nationally representative sample and applying a complex multistage probability sampling design. Second, this study systematically analyzed the association between BRI and COPD and adjusted for multiple covariates to minimize confounding. Moreover, regression and subgroup analyses further reinforced the reliability of the observed associations.

    Nevertheless, some limitations should be acknowledged. First, a causal relationship between BRI and COPD cannot be established because of the cross-sectional design. Second, since the data were derived from a US population, the generalizability of the findings is uncertain and warrants further validation. Third, compared with standardized diagnoses based on lung function tests, self-reported diagnoses may substantially underestimate the true prevalence of COPD, resulting in underdiagnosis. Consequently, self-report diagnosis is an imprecise tool for identifying COPD patients,45 potentially affecting the interpretation of the findings. Hence, these findings should be confirmed by prospective cohort studies.

    Conclusion

    This study observed a positive linear association between BRI and the likelihood of developing COPD. This result suggests that BRI could serve as a potential marker for assessing the likelihood of COPD and may help reduce the cost of early COPD screening. In addition, BRI has been shown to be more accurate and sensitive than BMI and WC. Therefore, these findings hold significant implications for clinical practice and epidemiological research. Future studies should further investigate whether BRI-based interventions can improve clinical outcomes in individuals with COPD, as well as explore the potential threshold value of BRI.

    Data Sharing Statement

    The datasets analysed during the current study are available in the National Center for Health Statistics (NCHS), https://www.cdc.gov/nchs/nhanes/about/index.html.

    Ethics Approval and Informed Consent

    All NHANES protocols obtained approval from the Ethics Review Board of NCHS. Written informed consent was offered by enrolled individuals.

    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

    The authors declare that they did not receive any funding from any source.

    Disclosure

    The authors declare that they have no competing interests.

    References

    1. Patel N. An update on COPD prevention, diagnosis, and management: the 2024 GOLD report. Nurse Pract. 2024;49:29–36. doi:10.1097/01.NPR.0000000000000180

    2. Sandelowsky H, Weinreich UM, Aarli BB, et al. COPD – do the right thing. BMC Fam Pract. 2021;22:244. doi:10.1186/s12875-021-01583-w

    3. James SL, Abate D, Abate KH, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet. 2018;392:1789–1858. doi:10.1016/S0140-6736(18)32279-7

    4. Wheaton AG, Liu Y, Croft JB, et al. Chronic obstructive pulmonary disease and smoking status – United States, 2017. MMWR Morb Mortal Wkly Rep. 2019;68:533–538. doi:10.15585/mmwr.mm6824a1

    5. Ho T, Cusack RP, Chaudhary N, et al. Under- and over-diagnosis of COPD: a global perspective. Breathe. 2019;15:24–35. doi:10.1183/20734735.0346-2018

    6. Martinez FJ, Han MK, Allinson JP, et al. At the root: defining and halting progression of early chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;197:1540–1551. doi:10.1164/rccm.201710-2028PP

    7. Palma G, Sorice GP, Genchi VA, et al. Adipose tissue inflammation and pulmonary dysfunction in obesity. Int J Mol Sci. 2022;23(13):7349. doi:10.3390/ijms23137349

    8. Furutate R, Ishii T, Wakabayashi R, et al. Excessive visceral fat accumulation in advanced chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2011;6:423–430. doi:10.2147/COPD.S22885

    9. Rahman I. Oxidative stress in pathogenesis of chronic obstructive pulmonary disease: cellular and molecular mechanisms. Cell Biochem Biophys. 2005;43:167–188. doi:10.1385/CBB:43:1:167

    10. Thomas DM, Bredlau C, Bosy-Westphal A, et al. Relationships between body roundness with body fat and visceral adipose tissue emerging from a new geometrical model. Obesity. 2013;21:2264–2271. doi:10.1002/oby.20408

    11. Huang C, Gao Z, Huang Z, et al. Nonlinear association between body roundness index and metabolic dysfunction associated steatotic liver disease in nondiabetic Japanese adults. Sci Rep. 2025;15:15442. doi:10.1038/s41598-025-99540-5

    12. Yang L, Huang S, Sheng S, et al. Association of obesity-related indices with rapid kidney function decline and chronic kidney disease, a study from a large longitudinal cohort in China. Obes Facts;2025. 1–27. doi:10.1159/000545356

    13. Wang K, Mao Y, Lu M, et al. Association between serum Klotho levels and the prevalence of diabetes among adults in the United States. Front Endocrinol. 2022;13:1005553. doi:10.3389/fendo.2022.1005553

    14. Cai Y, Chen M, Zhai W, et al. Interaction between trouble sleeping and depression on hypertension in the NHANES 2005–2018. BMC Public Health. 2022;22:481. doi:10.1186/s12889-022-12942-2

    15. Naik D, Joshi A, Paul TV, et al. Chronic obstructive pulmonary disease and the metabolic syndrome: consequences of a dual threat. Indian J Endocrinol Metab. 2014;18:608–616. doi:10.4103/2230-8210.139212

    16. Zhang Q, Wang Z, Liu W, et al. Elucidating the relationship between body fat index and pulmonary health: insights from cross-sectional analysis and mendelian randomization. Int J Chron Obstruct Pulmon Dis. 2025;20:869–882. doi:10.2147/COPD.S488523

    17. Verhulst SL, Schrauwen N, Haentjens D, et al. Sleep-disordered breathing in overweight and obese children and adolescents: prevalence, characteristics and the role of fat distribution. Arch Dis Child. 2007;92:205–208. doi:10.1136/adc.2006.101089

    18. Ji M, Zhang S, An R. Effectiveness of A Body Shape Index (ABSI) in predicting chronic diseases and mortality: a systematic review and meta-analysis. Obes Rev. 2018;19:737–759. doi:10.1111/obr.12666

    19. Song X, Jousilahti P, Stehouwer CD, et al. Comparison of various surrogate obesity indicators as predictors of cardiovascular mortality in four European populations. Eur J Clin Nutr. 2013;67:1298–1302. doi:10.1038/ejcn.2013.203

    20. Cao H, Shi C, Aihemaiti Z, et al. Association of body round index with chronic kidney disease: a population-based cross-sectional study from NHANES 1999–2018. Int Urol Nephrol. 2025;57:965–971. doi:10.1007/s11255-024-04275-3

    21. Wei C, Zhang G. Association between body roundness index (BRI) and gallstones: results of the 2017–2020 national health and nutrition examination survey (NHANES). BMC Gastroenterol. 2024;24:192. doi:10.1186/s12876-024-03280-1

    22. Qiu L, Xiao Z, Fan B, et al. Association of body roundness index with diabetes and prediabetes in US adults from NHANES 2007–2018: a cross-sectional study. Lipids Health Dis. 2024;23:252. doi:10.1186/s12944-024-02238-2

    23. Pan X, Liu F, Fan J, et al. Association of body roundness index and a body shape index with obstructive sleep apnea: insights from NHANES 2015–2018 data. Front Nutr. 2024;11:1492673. doi:10.3389/fnut.2024.1492673

    24. Xu J, Xiong J, Jiang X, et al. Association between body roundness index and weight-adjusted waist index with asthma prevalence among US adults: the NHANES cross-sectional study, 2005–2018. Sci Rep. 2025;15:9781. doi:10.1038/s41598-025-93604-2

    25. Leone N, Courbon D, Thomas F, et al. Lung function impairment and metabolic syndrome: the critical role of abdominal obesity. Am J Respir Crit Care Med. 2009;179:509–516. doi:10.1164/rccm.200807-1195OC

    26. Lam KB, Jordan RE, Jiang CQ, et al. Airflow obstruction and metabolic syndrome: the Guangzhou Biobank Cohort Study. Eur Respir J. 2010;35:317–323. doi:10.1183/09031936.00024709

    27. Lenártová P, Habánová M, Mrázová J, et al. Analysis of visceral fat in patients with chronic obstructive pulmonary disease (COPD). Rocz Panstw Zakl Hig. 2016;67:189–196.

    28. Huang JX, Xiao BJ, Yan YX, et al. Association between visceral adipose tissue and chronic respiratory diseases: a two-sample multivariable mendelian randomization study in European population. Int J Chron Obstruct Pulmon Dis. 2025;20:919–928. doi:10.2147/COPD.S510828

    29. Para O, Cassataro G, Fantoni C, et al. Prognostic role of blood eosinophils in acute exacerbations of chronic obstructive pulmonary disease: systematic review and meta-analysis. Monaldi Arch Chest Dis. 2025. doi:10.4081/monaldi.2025.3298

    30. Hughes MJ, McGettrick HM, Sapey E. Shared mechanisms of multimorbidity in COPD, atherosclerosis and type-2 diabetes: the neutrophil as a potential inflammatory target. Eur Respir Rev. 2020;29:190102. doi:10.1183/16000617.0102-2019

    31. Ren Y, Zhao H, Yin C, et al. Adipokines, hepatokines and myokines: focus on their role and molecular mechanisms in adipose tissue inflammation. Front Endocrinol. 2022;13:873699.

    32. Uchida K, Shiraishi M, Naito Y, et al. Activation of stress signaling pathways by the end product of lipid peroxidation. 4-hydroxy-2-nonenal is a potential inducer of intracellular peroxide production. J Biol Chem. 1999;274:2234–2242. doi:10.1074/jbc.274.4.2234

    33. Tsukagoshi H, Kawata T, Shimizu Y, et al. 4-Hydroxy-2-nonenal enhances fibronectin production by IMR-90 human lung fibroblasts partly via activation of epidermal growth factor receptor-linked extracellular signal-regulated kinase p44/42 pathway. Toxicol Appl Pharmacol. 2002;184:127–135. doi:10.1006/taap.2002.9514

    34. Suzuki M, Makita H, Östling J, et al. Lower leptin/adiponectin ratio and risk of rapid lung function decline in chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2014;11:1511–1519. doi:10.1513/AnnalsATS.201408-351OC

    35. Ruvuna L, Hijazi K, Guzman DE, et al. Dynamic and prognostic proteomic associations with FEV(1) decline in chronic obstructive pulmonary disease. medRxiv. 2024. doi:10.1101/2024.08.07.24311507

    36. Strain T, Wijndaele K, Sharp SJ, et al. Impact of follow-up time and analytical approaches to account for reverse causality on the association between physical activity and health outcomes in UK Biobank. Int J Epidemiol. 2020;49:162–172. doi:10.1093/ije/dyz212

    37. Willemse BW, ten Hacken NH, Rutgers B, et al. Effect of 1-year smoking cessation on airway inflammation in COPD and asymptomatic smokers. Eur Respir J. 2005;26:835–845. doi:10.1183/09031936.05.00108904

    38. Sahle BW, Chen W, Rawal LB, et al. Weight gain after smoking cessation and risk of major chronic diseases and mortality. JAMA Network Open. 2021;4:e217044. doi:10.1001/jamanetworkopen.2021.7044

    39. Saxton SN, Clark BJ, Withers SB, et al. Mechanistic links between obesity, diabetes, and blood pressure: role of perivascular adipose tissue. Physiol Rev. 2019;99(4):1701–1763. doi:10.1152/physrev.00034.2018

    40. Piché ME, Tchernof A, Després JP. Obesity phenotypes, diabetes, and cardiovascular diseases. Circ Res. 2020;126:1477–1500. doi:10.1161/CIRCRESAHA.120.316101

    41. Neeland IJ, Poirier P, Després JP. Cardiovascular and metabolic heterogeneity of obesity: clinical challenges and implications for management. Circulation. 2018;137:1391–1406. doi:10.1161/CIRCULATIONAHA.117.029617

    42. Nyberg M, Terzic D, Ludvigsen TP, et al. A state of natriuretic peptide deficiency. Endocr Rev. 2023;44:379–392. doi:10.1210/endrev/bnac029

    43. Power ML, Schulkin J. Sex differences in fat storage, fat metabolism, and the health risks from obesity: possible evolutionary origins. Br J Nutr. 2008;99:931–940. doi:10.1017/S0007114507853347

    44. Mafort TT, Rufino R, Costa CH, et al. Obesity: systemic and pulmonary complications, biochemical abnormalities, and impairment of lung function. Multidiscip Respir Med. 2016;11:28. doi:10.1186/s40248-016-0066-z

    45. Abrham Y, Zeng S, Lin W, et al. Self-report underestimates the frequency of the acute respiratory exacerbations of COPD but is associated with BAL neutrophilia and lymphocytosis: an observational study. BMC Pulm Med. 2024;24:433. doi:10.1186/s12890-024-03239-8

    Continue Reading

  • BNP Paribas sees retail banking recovery boosting capital generation through to 2028 – Reuters

    1. BNP Paribas sees retail banking recovery boosting capital generation through to 2028  Reuters
    2. France’s BNP to target 13% ROTE in 2028  Business Recorder
    3. BNP Paribas Targets Higher Profitability in 2028  The Wall Street Journal
    4. BNP Paribas Targets 13% ROTE by 2028 on Retail Banking Recovery: Trading Takeaways for EU Bank Stocks and Crypto (BTC, ETH)  Blockchain News
    5. France’s BNP sees retail banking recovery driving 13% ROTE in 2028  Global Banking | Finance | Review

    Continue Reading

  • ICC rejects Pakistan Cricket Board’s demand to remove match referee Andy Pycroft from Asia Cup 2025

    ICC rejects Pakistan Cricket Board’s demand to remove match referee Andy Pycroft from Asia Cup 2025

    The ICC on Tuesday rejected the Pakistan Cricket Board’s demand to remove match referee Andy Pycroft from the panel of officials for the ongoing Asia Cup.

    The PCB had filed a complaint with the ICC alleging that Pycroft asked Pakistan captain Salman Ali Agha, at the time of the toss, not to shake hands with his Indian counterpart Suryakumar Yadav in their Asia Cup match on Sunday.

    “Late last night, ICC had sent a reply to PCB stating that Pycroft won’t be removed and their plea has been rejected,” an ICC source told PTI.

    ALSO READ | India’s ODI future without Rohit and Kohli is no longer theoretical

    The 69-year-old Zimbabwean is due to officiate Pakistan’s final group stage game against the UAE on Wednesday.

    Pakistan team manager Naved Cheema had also filed a complaint with the Asian Cricket Council, alleging that it was on Pycroft’s insistence that team sheets weren’t exchanged between the two skippers on Sunday, as is the norm.

    Published on Sep 16, 2025

    Continue Reading

  • Pollo AI Launches Seedream 4.0 With 60 Free Generations for All Paid Users

    SINGAPORE, Sept. 16, 2025 /PRNewswire/ — Pollo AI is excited to announce the official integration of Seedream 4.0, the latest AI image model from ByteDance’s Seedream AI. Starting today, all paid subscribers get 60 FREE generations within 5 days to experience it firsthand, available for both text to image and image to image creations, at no additional cost.

    Seedream 4.0 sets a new standard in AI image generation with its ability to deliver consistent results and maintain visual cohesion across multiple outputs.

    For image to image, users can upload up to 10 source images and blend them into a single, cohesive image. This maintains subject/character consistency throughout the composition, perfect for creating consistent characters, product shots, architectural renders, or image series with unified style.

    “Whether for marketing campaigns, branding, or storytelling, Seedream 4.0 ensures professional-grade output every time.”, Emma Chen, Pollo AI’s Chief Product Officer, shared.

    Now, all paid users of Pollo AI get immediate access to this powerful model along with the special offer of 60 FREE generations within the first 5 days. Don’t miss this chance to unlock advanced AI image generation capabilities.

    Getting started is simple. All you need to do is go to Pollo AI image generator and pick Seedream 4.0 as the generation model! Don’t wait! The offer ends soon.

    Media Contact
    Emma Chen
    Chief Product Officer, Pollo AI
    [email protected] 

    SOURCE Pollo AI

    Continue Reading

  • The associations of neutrophil-to-lymphocyte ratio, monocyte-to-lympho

    The associations of neutrophil-to-lymphocyte ratio, monocyte-to-lympho

    Introduction

    Glial fibrillary acidic protein (GFAP) is an intermediate filament protein in astrocytes and an important component of the astrocyte cytoskeleton, which plays an important role in astrocyte regeneration, synaptic plasticity, and reactive gliosis.1,2 Abnormal regulation and expression of GFAP is related to various neurological disorders, including infectious diseases of the central nervous system (CNS), neurodegenerative diseases, cerebral edema, traumatic brain injury, and psychiatric disorders.3 In 2016, the United States’ Mayo Clinic Lennon team identified anti-GFAP antibodies in the cerebrospinal fluid and serum of patients with meningoencephalomyelitis and named it autoimmune GFAP astrocytopathy (GFAP-A).4 GFAP-A affects the brain, meninges, spinal cord, and/or optic nerve. Its clinical manifestations are complex and varied, including fever, headache, ataxia, seizures, mental and behavioral abnormalities, movement disorders, tremors, and autonomic and cognitive dysfunction.5,6 In addition, due to the heterogeneity of the disease, GFAP-A may become life-threatening within weeks or even days in some patients due to central hypoventilation or severe autonomic dysfunction.6–9 Currently, there are no unified standards or guidelines for the treatment of GFAP-A. Most patients with GFAP-A respond well to steroid therapy and achieve complete or partial remission upon discharge, and a favorable prognosis. However, some patients are prone to recurrence, leaving varying degrees of functional disability and even death.5,6,8,10 Therefore, it is vital to establish biomarkers to assess appropriate steroid therapy and clinical treatment decisions for GFAP-A.

    Monitoring disease progression and detecting potentially severe disease at an early stage are crucial for managing individual patients with GFAP-A, as this affects clinical treatment decisions. Studies focused on biomarkers related to disease severity and prognosis of GFAP-A are in the beginning stages. Segal et al11 found that higher neurofilament light chain (NfL) concentrations were associated with magnetic resonance imaging abnormalities and a poor prognosis in patients with GFAP-A, suggesting that NfL may be a biomarker of disease severity and prognosis in GFAP-A. Fu et al12 found that MIP-3α was positively correlated with disease severity by detecting the expression levels of 200 serum cytokines in patients with GFAP-A. Kimura et al13 observed elevated levels of TNF-α, IL-27, IL-6, CCL20, GFAP, S100 calcium-binding protein B, and NfL in GFAP-A cerebrospinal fluid samples, suggesting that these may be potential biomarkers of GFAP-A. However, these potential biomarkers of GFAP-A have some limitations in clinical practice. First, due to economic considerations, not all patients will undergo testing for the above biological indicators. Second, due to the requirements of the detection methods, not all hospitals are capable of conducting the detection of the above biological indicators. Finally, cytokines and chemokines in cerebrospinal fluid may be degraded during storage, resulting in inaccurate test results. Therefore, there is an urgent need for objective, inexpensive, and clinically accessible biomarkers to guide the clinical management of patients with GFAP-A.

    Pathological studies have revealed the extensive infiltration of lymphocytes, monocytes, and neutrophils around intracranial blood vessels in patients with GFAP-A, which is consistent with the typical imaging feature of GFAP-A with perivascular linear radiolucent enhancement perpendicular to the ventricle of the brain.6,14–16 These findings suggest that neuroinflammation caused by the interaction of lymphocytes, monocytes, neutrophils, microglia, and antibodies secreted by plasma cells is a possible pathogenesis of GFAP-A. Neutrophils, monocytes, and lymphocytes are inflammatory markers measured using routine blood examinations that are often used to reflect the inflammatory status in patients. The neutrophil-to-lymphocyte ratio (NLR) and monocyte-to-lymphocyte ratio (MLR) are more representative of the inflammatory state of the body than neutrophil, monocyte, or lymphocyte counts alone. Studies have observed that the NLR and MLR are associated with the severity, activity, and prognosis of some immune diseases of the CNS, including multiple sclerosis (MS), autoimmune encephalitis (AE), neuromyelitis optica spectrum disorder (NMOSD), and myelin-oligodendrocyte glycoprotein antibody-associated disease (MOGAD).17–19 Similar to the NLR and MLR, the platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammatory index (SII) are associated with the severity, activity, and prognosis of several immune diseases, including NMOSD, MOGAD, and AE.19–21 However, the associations of the NLR, MLR, PLR, and SII with the disease severity and prognosis of patients with GFAP-A remain unclear. This study analyzed the associations between these inflammatory indices and the disease severity and prognosis of patients with GFAP-A.

    Materials and Methods

    Study Patients

    Sixty-two patients with GFAP-A who were treated at the First Affiliated Hospital of Zhengzhou University between January 2020 and August 2024 were included in this study (Figure 1). The age range of these patients was 3 to 68 years old. This study followed the principles of the Declaration of Helsinki and was approved by the Ethics Committee of the First Affiliated Hospital of Zhengzhou University (No. 2022-KY-0053). All patients included in this study were newly diagnosed with GFAP-A. Currently, there is no unified standard for the diagnosis of GFAP-A. Some patients are often misdiagnosed as viral encephalitis, tuberculous meningitis, or acute disseminated encephalomyelitis, leading to delayed and incorrect treatment.22,23 We have summarized the following diagnostic points based on relevant literature: (1) acute or subacute onset; (2) clinical manifestations of meningeal, cerebral, spinal cord, or optic nerve involvement or a combination of symptoms; (3) paraventricular linear radial enhancement and/or spinal cord long-segment involvement on MRI; (4) serum and/or cerebrospinal fluid anti-GFAP antibody positivity; (5) responsive to steroid hormone therapy; (6) the exclusion of other diagnoses.4–6,14 In addition, all blood samples used in this study were obtained prior to the administration of steroid hormones or immunosuppressants, and all relevant clinical data were available for all patients in this study. Patients were excluded from this study if it was not their first GFAP-A attack or they had comorbidities of a tumor, rheumatism, infectious diseases, hematological diseases, other autoimmune diseases, or psychiatric disorders. Patients with severe liver, kidney, or other organic diseases, or other autoimmune antibodies, such as anti-AQP4 antibodies or anti-MOG antibodies, were also excluded from this study. Pregnant patients, those who had been administered steroid hormones or immunosuppressants prior to blood collection, and those with antiplatelet drug therapy use (including aspirin and/or clopidogrel) within 4 months of the study were excluded. Finally, patients with incomplete clinical information were also excluded from this study.

    Figure 1 Flowchart of included and excluded patients.

    Data Collection

    Basic clinical data, laboratory findings, and imaging data were collected from all of the patients. Basic clinical data included sex, age, prodromal events, time from first symptom to consultation, time from first symptom to diagnosis, need for mechanical ventilation, need for intensive care unit (ICU) treatment for at least 48h, clinical manifestations, immunotherapy, and length of hospitalization. Laboratory findings included routine blood examination results, liver function, kidney function, rheumatological and immune indices, tumor markers, cerebrospinal fluid results, and serum and/or cerebrospinal fluid antibody test results. Routine blood examinations were conducted within 24 hours of admission and before immunotherapy in all patients during acute attacks (episodes of neurological deterioration lasting more than 24 hours). Routine blood examination results included white blood cell, platelet, neutrophil, monocyte, lymphocyte, eosinophil, basophil counts. The NLR, MLR, PLR, and SII were calculated based on the routine blood examination results (NLR=neutrophils/lymphocytes; MLR=monocytes/lymphocytes; PLR=platelets/lymphocytes; SII=neutrophils*platelets/lymphocytes). Long-term follow-up was conducted after immunotherapy.

    Assessment Scale

    The primary endpoints of this study were disease severity at admission and prognosis. The Clinical Assessment Scale for Autoimmune Encephalitis (CASE) consists of nine items, including seizure, memory dysfunction, psychiatric symptoms, consciousness, language problem, dyskinesia/dystonia, gait instability and ataxia, brainstem dysfunction, and weakness, which compensates for the shortcomings of the modified Rankin Scale (mRS) in assessing non-motor symptoms of autoimmune encephalitis.24 GFAP-A mainly manifests as a combination of the above symptoms; therefore, CASE was used to assess the disease severity of GFAP-A at admission, and patients were categorized based on the CASE score into mild (CASE ≤4) or severe (CASE ≥5) groups. The mRS was used to assess the patients’ conditions at discharge and follow-up, and patients were categorized based on the mRS score at 1-year follow-up (one year after discharge) into the good (mRS ≤2) or poor (mRS ≥3) prognosis groups.

    Statistical Analysis

    All statistical analyses were conducted using SPSS statistical software (version 25.0; IBM, Chicago, IL, USA). Continuous data with normal distribution were presented as mean ± standard deviation, and an independent samples t-test was used to compare the continuous data. Continuous data that did not conform to a normal distribution were presented as median and interquartile range, and the Mann–Whitney U-test was used to compare the continuous data. Categorical data were compared using the chi-square test or Fisher’s exact probability method. Spearman correlation analysis was used to test the correlations between the NLR, MLR, PLR, and SII and the severity of GFAP-A. Receiver operating characteristic (ROC) curves were used to evaluate the ability of the NLR, MLR, PLR, and SII to predict the severity of GFAP-A, and the area under the curve (AUC) was calculated. Binary logistic regression was used to analyze the risk factors affecting the disease severity and prognosis of patients with GFAP-A. This study only included patients with GFAP-A and did not compare the GFAP-A groups with healthy controls or other patient groups. All statistical analyses were two-sided, and the level of statistical significance was set at P < 0.05.

    Results

    Patient Characteristics

    Sixty-two patients with GFAP-A were included in this study. The clinical features and laboratory findings are shown in Table 1. The clinical manifestations of the patients in this study were diverse and included fever (n=43, 69.35%), headache (n=32, 53.33%), ataxia (n=22, 35.48%), movement disorders (n=15, 24.19%), cognitive dysfunction (n=11, 17.74%), mental and behavioral abnormalities (n=6, 9.68%), seizures (n=5, 8.06%), consciousness disorders (n=17, 27.42%), autonomic dysfunction (n=34, 54.84%), tremors (n=9, 14.52%), and visual abnormalities (n=10, 16.13%). All patients were administered first-line immunotherapy, and no patient received second-line immunotherapy. Fifty-five patients were administered oral steroid hormones and/or mycophenolate mofetil after discharge.

    Table 1 Clinical Features and Laboratory Findings of Patients with GFAP-A

    The NLR Was Associated with the Severity of GFAP-A

    Clinical Data of Patients in the Mild and Severe Groups

    The mild group included 42 patients (67.74%), and the severe group included 20 patients (32.26%). The neutrophil count, NLR, MLR, PLR, and SII were significantly higher in patients in the severe group than in those in the mild group (P <0.05). The basophil count was significantly lower in patients in the severe group than in those in the mild group (P <0.05). No significant differences were observed between the two groups in terms of sex, age, prodromal events, time from first symptom to consultation, and white blood cell, platelet, monocyte, lymphocyte, or eosinophil counts (Table 2).

    Table 2 Clinical Data of Patients in the Mild and Severe Groups

    Correlations of the NLR, MLR, PLR, and SII with the Severity of GFAP-A

    As shown in Figure 2, the NLR, MLR, and SII were positively correlated with the CASE score of patients with GFAP-A (r=0.365, P=0.003; r=0.283, P=0.026; r=0.313, P=0.013), while no significant correlation was observed between the PLR and the CASE score (r=0.236, P=0.064).

    Figure 2 Correlations of the NLR, MLR, PLR, and SII with the CASE score (ad).

    Abbreviations: NLR, neutrophil-to-lymphocyte ratio; MLR, monocyte-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; SII, systemic immune-inflammatory index; CASE, the Clinical Assessment Scale for Autoimmune Encephalitis.

    The Predictive Value of the NLR, MLR, PLR, and SII for the Severity of GFAP-A

    Figure 3 showed ROC curves of the NLR, MLR, PLR, and SII to predict the severity of GFAP-A. The optimal cut-off value of the NLR for predicting the severity of GFAP-A was 3.05, with a sensitivity of 80%, specificity of 57.1%, and AUC of 0.729 (95% Confidence Interval/CI: 0.600–0.858, P=0.004). The optimal cut-off value of the MLR for predicting the severity of GFAP-A was 0.35, with a sensitivity of 65%, specificity of 66.7%, and AUC of 0.66 (95% CI: 0.512–0.807, P=0.044). The optimal cut-off value of the PLR for predicting the severity of GFAP-A was 176.34, with a sensitivity of 65%, specificity of 73.8%, and AUC of 0.656 (95% CI: 0.504–0.808, P=0.049). The optimal cut-off value of the SII for predicting the severity of GFAP-A was 1055.43, with a sensitivity of 60%, specificity of 76.2%, and AUC of 0.702 (95% CI: 0.563–0.841, P=0.01). Details of the AUC, optimal cut-off, sensitivity, and specificity were shown in Table 3.

    Table 3 The Predictive Value of the NLR, MLR, PLR, and SII for the Severity of GFAP-A

    Figure 3 ROC curves of the NLR, MLR, PLR, and SII for predicting the severity of GFAP-A.

    Abbreviations: ROC, receiver operating characteristic; GFAP-A, autoimmune glial fibrillary acidic protein astrocytopathy; NLR, neutrophil-to-lymphocyte ratio; MLR, monocyte-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; SII, systemic immune-inflammatory index.

    The NLR as an Independent Risk Factor for the Severity of GFAP-A

    The neutrophil, lymphocyte, and basophil counts, and the NLR, MLR, and SII were correlated with the GFAP-A severity (P <0.05) (Table 4). The NLR (Odds Ratio/OR=1.238, 95% CI: 1.003–1.473, P=0.01) was identified as an independent risk factor for the severity of GFAP-A (Table 4).

    Table 4 Binary Logistic Regression Analysis of Factors Related to the Severity of GFAP-A

    The mRS Score at Discharge as an Independent Risk Factor for the Poor Prognosis One year After Discharge of GFAP-A

    Among the 62 patients included in this study, 12 were followed up for less than 1 year, and two were lost to follow-up; therefore, these 14 patients were excluded from the follow-up analysis. The remaining 48 patients were grouped according to the mRS score at 1-year follow-up (one year after discharge). Forty-one patients were included in the good prognosis group (85.42%) and seven patients were included in the poor prognosis group (14.58%). The need for mechanical ventilation, ICU treatment, length of hospitalization, CASE score at admission and at the worst condition, and mRS score at discharge were associated with a poor prognosis one year after discharge (P <0.05) (Table 5). The mRS score at discharge (OR=7.966, 95% CI: 1.120–56.658; P=0.038) was identified as an independent risk factor for the poor prognosis one year after discharge of patients with GFAP-A (Table 6).

    Table 5 Univariate Logistic Regression Analysis of Factors Associated with a Poor Prognosis in GFAP-A

    Table 6 Multivariate Logistic Regression Analysis of Factors Associated with a Poor Prognosis in GFAP-A

    Changes in the NLR, MLR, PLR, and SII After Administration of Immunotherapy

    Routine blood examination results during follow-up were available for 32 patients, including 27 with a good prognosis and five with a poor prognosis. The NLR, MLR, and SII were not significantly different before and after immunotherapy (P >0.05); however, the PLR was significantly reduced after immunotherapy (153.01 vs 121.16, P=0.039) (Table 7). Twenty-four patients had a reduced PLR after immunotherapy. However, a decrease in the PLR after immunotherapy was not correlated with the prognosis of patients with GFAP-A (P=0.779).

    Table 7 The NLR, MLR, PLR, and SII Before and After Immunotherapy in GFAP-A

    Discussion

    GFAP-A is a novel autoimmune inflammatory disease of the CNS associated with antibodies to GFAP. Meningitis, encephalitis, myelitis, optic neuritis, or a combination of these serve as the main clinical manifestation, and the typical imaging feature is perivascular linear radiolucent enhancement perpendicular to the ventricle of the brain.5,6 Most patients with GFAP-A respond well to treatment such as steroid hormones, intravenous immunoglobulin, plasma exchange, immunosuppressants, and monoclonal antibodies and have a good prognosis; however, some patients relapse, and the disease may progress to residual dysfunction or death.6,25 In the current study, the majority of patients with GFAP-A were middle-aged, and the number of male patients was higher than the number of female patients. These findings differ from those of previous studies, in which no difference in sex was reported among patients with GFAP-A.4,6,22 These differing results may be related to racial and economic differences or a result of the insufficient sample size in the current study. Similar to previous studies, patients with GFAP-A presented with fever, headache, ataxia, movement disorders, and autonomic dysfunction in this study.4,6 Despite the gradual increase in the number of studies regarding GFAP-A, few studies regarding biological indicators related to the disease severity and patient prognosis have been reported. Objective and inexpensive biological indicators are urgently needed to guide clinical practice.

    Neuroinflammation caused by interactions between lymphocytes, monocytes, neutrophils, microglia, and antibodies secreted by plasma cells may be a pathogenetic mechanism of GFAP-A.6,14–16 Long et al14 retrospectively analyzed 19 patients with GFAP-A, including four who underwent brain tissue biopsy. The neuropathological results suggested that the inflammatory lesions were mainly infiltrated by T lymphocytes, B lymphocytes, plasma cells, scattered neutrophils, and eosinophils with obvious activation of microglia around the cerebral blood vessels.14 Yamakawa et al26 reviewed the neuropathological results at autopsy in two patients with GFAP-A, and reported findings consistent with those reported by Long et al. Iorio et al15 observed inflammatory changes in the local meningeal tissue infiltrated by macrophages and CD8+ T cells in a meningeal biopsy specimen of a patient with GFAP-A. Neutrophils and monocytes play important roles in the innate immune system. Neutrophils can damage the blood-brain barrier and increase its permeability by releasing a variety of proinflammatory factors, thus promoting the development of autoimmune diseases of the CNS.27–29 Moreover, neutrophils can induce monocytes to accumulate at the site of inflammation by releasing large amounts of cytokines. Subsequently, monocytes differentiate into macrophages, which play an important role in the formation of antigen-presenting cells and activate lymphocytes to initiate acquired immunity.28,30,31 T and B lymphocytes are vital members of acquired immunity, and cytotoxic CD8+ T cell-mediated immune responses play an important role in the pathogenesis of GFAP-A.4,32 In addition, it has been reported that platelets can transmit signals for leukocyte differentiation, migration, and infiltration via the secretion of cytokines, chemokines, and their receptors, thus playing an important role in inflammatory diseases of the CNS, such as MS and MOGAD.33–36 Platelets can increase the permeability of the blood-brain barrier by promoting neutrophil migration, thereby promoting the progression of neuroinflammation.37,38 Neutrophils, monocytes, lymphocytes, and platelets are biological indicators derived from routine blood examinations that reflect the inflammatory state of the body. Routine blood examinations are easily performed, clinically accessible, and inexpensive, and can provide a detailed snapshot of the peripheral immune cells. Currently, a single leukocyte subtype count is commonly used for the clinical assessment of the degree of inflammation in the body, which is susceptible to physiological and/or pathological factors. In contrast, the NLR, MLR, PLR, and SII have higher stability and can more accurately assess the inflammatory degree in the body and reflect disease severity.39

    Monitoring disease progression and the early detection of potentially severe disease are critical for the management of individual patients with GFAP-A, as it can help determine the optimal treatment, such as the need for combination first-line immunotherapy, early use of monoclonal antibodies, or addition of immunosuppressants. However, there is currently no ideal biomarker for predicting the severity and progression of GFAP-A. The NLR, MLR, PLR, and SII are related to the severity, activity, and prognosis of several autoimmune diseases of the CNS. A study of 199 patients with AE reported that the NLR and MLR were significantly increased in patients with severe AE, and a high NLR and MLR were independent risk factors for patients with severe AE, though they were not associated with patient prognosis.17 In contrast to the conclusions of Liu et al, Qiu et al40 found that a high NLR on admission was correlated with a poor prognosis in patients with AE, which may be due to differences in research methods, sample size, and follow-up times between the studies. Hemond et al41 found that the NLR and MLR were closely associated with the severity of neurological dysfunction and whole-brain atrophy in patients with MS, and that patients with a high NLR were more likely to relapse.42 Lin et al19 reported that a high PLR level was positively correlated with MOGAD activity and relapse and that the PLR can be used to differentiate between MS and NMOSD.43 Moreover, Mao et al21 reported that the SII was related to the disease severity and prognosis of patients with AE. No studies regarding the associations of the NLR, MLR, PLR, and SII with GFAP-A have been reported. Therefore, this is the first study to investigate the associations between these inflammatory indices and the severity and prognosis of GFAP-A. In the current study, the NLR (OR=1.238, 95% CI: 1.003–1.473, P=0.01) was identified as an independent risk factor for the severity of GFAP-A and was positively correlated with the CASE score (r=0.365, P=0.003). The optimal cut-off value of the NLR was 3.05, with a sensitivity of 80%, specificity of 57.1%, and AUC of 0.729 (95% CI: 0.600–0.858, P=0.004), which supported the use of the NLR as a predictor of GFAP-A severity. In addition, the NLR, MLR, PLR, and SII measured at admission were not related to the prognosis of GFAP-A (P>0.05). The PLR significantly decreased after immunotherapy (P=0.039), though the reduction in the PLR after immunotherapy was not correlated with the prognosis of GFAP-A (P>0.05). Last, the mRS score at discharge (OR=7.966, 95% CI: 1.120–56.658; P=0.038) was identified as an independent risk factor for the poor prognosis one year after discharge of patients with GFAP-A. These findings are still in the preliminary stages and require external validation in the future. Our data suggest that an NLR of 3.05 or higher may indicate a severe condition of GFAP-A. For these patients, physicians should optimize clinical treatment decisions as early as possible, such as extending the duration of steroid therapy, combining first-line immunotherapy, or starting second-line immunotherapy early, which may reduce the likelihood of recurrence or residual neurological dysfunction in patients. However, we have not evaluated the correlation between NLR levels and the specific methods and duration of immunotherapy of the patients, and we will further investigate this correlation in the future. Taken together, these results indicate that the NLR is a potential biomarker to predict the severity of GFAP-A and monitor disease progression.

    This study is not without limitations. First, it was a small-sample and single-center study, rendering it susceptible to selection bias. Second, this was a retrospective study. The presented data were retrospectively obtained from an electronic medical record system, and the follow-up of most patients was conducted retrospectively, which may lead to information and confounding biases. Therefore, a prospective, multicenter study with a larger sample size is needed to verify the conclusions of the current study. Finally, since tumors, infectious diseases, hematological disorders, and other autoimmune diseases can affect NLR, MLR, PLR, and SII, we excluded patients with these comorbidities. Therefore, the results of this study may not be extrapolated to all patients with GFAP-A.

    Conclusion

    This is the first study to explore the associations between the NLR and GFAP-A severity. The NLR is a clinically inexpensive, accessible, and widely-available inflammatory marker that is positively associated with the severity of GFAP-A. Therefore, this biomarker can help physicians monitor disease progression and identify patients with severe disease at an early stage, thus allowing for optimal treatment decisions.

    Data Sharing Statement

    The data that support the results of this study are available from the corresponding author on reasonable request.

    Ethics Approval and Informed Consent

    This study was performed in line with the principles of the Declaration of Helsinki and approved by the Ethics Committee of the First Affiliated Hospital of Zhengzhou University (No. 2022-KY-0053). Informed consent was obtained from all individual participants included in the 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 was supported by Henan Province Science and Technology Tackle Project (grant number: 252102310280), the Outstanding Young Talent Cultivation Project of Henan Science and Technology Innovation Talents (grant number: YXKC2022037), and Young and Middle-aged Academic Leaders of Henan Provincial Health Commission.

    Disclosure

    All authors declare no conflicts of interest in this work.

    References

    1. Li D, Liu X, Liu T, et al. Neurochemical regulation of the expression and function of glial fibrillary acidic protein in astrocytes. Glia. 2019;68(5):878–897. doi:10.1002/glia.23734

    2. McKeon A, Benarroch EE. Glial fibrillary acid protein:functions and involvement in disease. Neurology. 2018;90(20):925–930. doi:10.1212/wnl.0000000000005534

    3. Hol EM, Pekny M. Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system. Curr Opin Cell Biol. 2015;32:121–130. doi:10.1016/j.ceb.2015.02.004

    4. Fang B, McKeon A, Hinson SR, et al. Autoimmune glial fibrillary acidic protein astrocytopathy:a novel meningoencephalomyelitis. JAMA Neurol. 2016;73(11). doi:10.1001/jamaneurol.2016.2549

    5. Kunchok A, Zekeridou A, McKeon A. Autoimmune glial fibrillary acidic protein astrocytopathy. Curr Opinion Neurol. 2019;32(3):452–458. doi:10.1097/wco.0000000000000676

    6. Flanagan EP, Hinson SR, Lennon VA, et al. Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: analysis of 102 patients. Ann Neurol. 2017;81(2):298–309. doi:10.1002/ana.24881

    7. Sommer S, Panzer A, Bertolini A, et al. Spectrum of clinical and imaging features of children with GFAP astrocytopathy. Neurol Neuroimmunol Neuroinflammation. 2025;12(1). doi:10.1212/nxi.0000000000200327

    8. Arzalluz-Luque J, Dumez P, Picard G, et al. Clinical course and long-term outcomes in autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy. J Neurol. 2025;272(6). doi:10.1007/s00415-025-13159-0

    9. Li J, Shen C, Chen Y, Zeng H, Cui L, Feng H. Organ donation after brain death from autoimmune glial fibrillary acidic protein astrocytopathy: a case report. Heliyon. 2024;10(7). doi:10.1016/j.heliyon.2024.e28558

    10. Hagbohm C, Ouellette R, Flanagan EP, et al. Clinical and neuroimaging phenotypes of autoimmune glial fibrillary acidic protein astrocytopathy: a systematic review and meta‐analysis. Eur J Neurol. 2024;31(7). doi:10.1111/ene.16284

    11. Segal Y, Mangioris G, Lennon V, et al. CSF cytokine, chemokine and injury biomarker profile of glial fibrillary acidic protein (GFAP) autoimmunity. Ann Clin Transl Neurol. 2025;12(4):855–860. doi:10.1002/acn3.52305

    12. Fu C-C, Huang L, Xu L-F, et al. Serological biomarkers in autoimmune GFAP astrocytopathy. Front Immunol. 2022;13. doi:10.3389/fimmu.2022.957361

    13. Kimura A, Takemura M, Yamamoto Y, Hayashi Y, Saito K, Shimohata T. Cytokines and biological markers in autoimmune GFAP astrocytopathy: the potential role for pathogenesis and therapeutic implications. J Neuroimmunol. 2019;334:576999. doi:10.1016/j.jneuroim.2019.576999

    14. Long Y, Liang J, Xu H, et al. Autoimmune glial fibrillary acidic protein astrocytopathy in Chinese patients: a retrospective study. Eur J Neurol. 2018;25(3):477–483. doi:10.1111/ene.13531

    15. Iorio R, Damato V, Evoli A, et al. Clinical and immunological characteristics of the spectrum of GFAP autoimmunity: a case series of 22 patients. J Neurol Neurosurg. 2018;89(2):138–146. doi:10.1136/jnnp-2017-316583

    16. Shu Y, Long Y, Chang Y, et al. Brain immunohistopathology in a patient with autoimmune glial fibrillary acidic protein astrocytopathy. Neuroimmunomodulation. 2018;25(1):1–6. doi:10.1159/000488879

    17. Liu Z, Li Y, Wang Y, Zhang H, Lian Y, Cheng X. The neutrophil-to-lymphocyte and monocyte-to-lymphocyte ratios are independently associated with the severity of autoimmune encephalitis. Front Immunol. 2022;13. doi:10.3389/fimmu.2022.911779

    18. Devlin L, Gombolay G. The neutrophil-to-lymphocyte ratio and the monocyte-to-lymphocyte ratio predict expanded disability status scale score at one year in pediatric neuromyelitis optica spectrum disorder but not in multiple sclerosis. Pediatr Neurol. 2023;143:84–88. doi:10.1016/j.pediatrneurol.2023.03.009

    19. Lin L, Ji M, Wu Y, Hang H, Lu J. Neutrophil to lymphocyte ratio may be a useful marker in distinguishing MOGAD and MS and platelet to lymphocyte ratio associated with MOGAD activity. Mult Scler Relat Disord. 2023;71:104570. doi:10.1016/j.msard.2023.104570

    20. Yan H, Wang Y, Li Y, et al. Combined platelet-to-lymphocyte ratio and blood-brain barrier biomarkers as indicators of disability in acute neuromyelitis optica spectrum disorder. Neurol Sci. 2023;45(2):709–718. doi:10.1007/s10072-023-07058-3

    21. Mao C, Cui X, Zhang S. The value of the systemic immune-inflammation index in assessing disease severity in autoimmune encephalitis. Int J Neurosci. 2024;1–8. doi:10.1080/00207454.2024.2410033

    22. Bai R, An L, Du W, et al. Autoimmune glial fibrillary acidic protein astrocytopathy misdiagnosed as intracranial infectious diseases: case reports and literature review. Front Immunol. 2025;16. doi:10.3389/fimmu.2025.1519700

    23. Li J, Wang C, Cao Y, et al. Autoimmune glial fibrillary acidic protein astrocytopathy mimicking acute disseminated encephalomyelitis. Medicine. 2021;100(25). doi:10.1097/md.0000000000026448

    24. Lim JA, Lee ST, Moon J, et al. Development of the clinical assessment scale in autoimmune encephalitis. Ann Neurol. 2019;85(3):352–358. doi:10.1002/ana.25421

    25. Zhang W, Xie Y, Wang Y, et al. Clinical characteristics and prognostic factors for short-term outcomes of autoimmune glial fibrillary acidic protein astrocytopathy: a retrospective analysis of 33 patients. Front Immunol. 2023;14. doi:10.3389/fimmu.2023.1136955

    26. Yamakawa M, Hogan KO, Leever J, Jassam YN. Autopsy case of meningoencephalomyelitis associated with glial fibrillary acidic protein antibody. Neurol Neuroimmunol Neuroinflammation. 2021;8(6). doi:10.1212/nxi.0000000000001081

    27. De Bondt M, Hellings N, Opdenakker G, Struyf S. Neutrophils: underestimated players in the pathogenesis of Multiple Sclerosis (MS). Int J Mol Sci. 2020;21(12):4558. doi:10.3390/ijms21124558

    28. Wesselingh R, Butzkueven H, Buzzard K, Tarlinton D, O’Brien TJ, Monif M. Innate immunity in the central nervous system: a missing piece of the autoimmune encephalitis puzzle? Front Immunol. 2019;10. doi:10.3389/fimmu.2019.02066

    29. Winkler A, Wrzos C, Haberl M, et al. Blood-brain barrier resealing in neuromyelitis optica occurs independently of astrocyte regeneration. J Clin Investig. 2021;131(5). doi:10.1172/jci141694

    30. Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011;11(8):519–531. doi:10.1038/nri3024

    31. Navegantes KC, de Souza Gomes R, Pereira PAT, Czaikoski PG, Azevedo CHM, Monteiro MC. Immune modulation of some autoimmune diseases: the critical role of macrophages and neutrophils in the innate and adaptive immunity. J Transl Med. 2017;15(1). doi:10.1186/s12967-017-1141-8

    32. Guo Y, Endmayr V, Zekeridou A, et al. New insights into neuropathology and pathogenesis of autoimmune glial fibrillary acidic protein meningoencephalomyelitis. Acta Neuropathol. 2024;147(1). doi:10.1007/s00401-023-02678-7

    33. Morrell CN, Aggrey AA, Chapman LM, Modjeski KL. Emerging roles for platelets as immune and inflammatory cells. Blood. 2014;123(18):2759–2767. doi:10.1182/blood-2013-11-462432

    34. Liu Y, Shields LBE, Gao Z, et al. Current understanding of platelet-activating factor signaling in central nervous system diseases. Mol Neurobiol. 2016;54(7):5563–5572. doi:10.1007/s12035-016-0062-5

    35. Langer HF, Choi EY, Zhou H, et al. Platelets contribute to the pathogenesis of experimental autoimmune encephalomyelitis. Circul Res. 2012;110(9):1202–1210. doi:10.1161/circresaha.111.256370

    36. Starossom SC, Veremeyko T, Yung AWY, et al. Platelets play differential role during the initiation and progression of autoimmune neuroinflammation. Circul Res. 2015;117(9):779–792. doi:10.1161/circresaha.115.306847

    37. Chou M-L, Babamale AO, Walker TL, Cognasse F, Blum D, Burnouf T. Blood–brain crosstalk: the roles of neutrophils, platelets, and neutrophil extracellular traps in neuropathologies. Trends Neurosci. 2023;46(9):764–779. doi:10.1016/j.tins.2023.06.005

    38. Chang BL, Ro LS, Chen CM, et al. Serum levels of cell adhesion molecules in patients with neuromyelitis optica spectrum disorder. Ann Clin Transl Neurol. 2020;7(10):1854–1861. doi:10.1002/acn3.51167

    39. Luo H, He L, Zhang G, et al. Normal reference intervals of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, and systemic immune inflammation index in healthy adults: a large multi-center study from Western China. Clin Lab. 2019;65(3). doi:10.7754/Clin.Lab.2018.180715

    40. Qiu X, Zhang H, Li D, et al. Analysis of clinical characteristics and poor prognostic predictors in patients with an initial diagnosis of autoimmune encephalitis. Front Immunol. 2019;10. doi:10.3389/fimmu.2019.01286

    41. Hemond CC, Glanz BI, Bakshi R, Chitnis T, Healy BC. The neutrophil-to-lymphocyte and monocyte-to-lymphocyte ratios are independently associated with neurological disability and brain atrophy in multiple sclerosis. BMC Neurol. 2019;19(1). doi:10.1186/s12883-019-1245-2

    42. Bisgaard AK, Pihl-Jensen G, Frederiksen JL. The neutrophil-to-lymphocyte ratio as disease activity marker in multiple sclerosis and optic neuritis. Mult Scler Relat Disord. 2017;18:213–217. doi:10.1016/j.msard.2017.10.009

    43. Carnero Contentti E, López PA, Criniti J, et al. Platelet-to-lymphocyte ratio differs between MS and NMOSD at disease onset and predict disability. Mult Scler Relat Disord. 2022:58. doi:10.1016/j.msard.2022.103507

    Continue Reading

  • How to protect your heart: Cardiologist shares 5 daily habits to lower your heart attack risk

    How to protect your heart: Cardiologist shares 5 daily habits to lower your heart attack risk

    Heart disease is a serious problem that leads to many deaths worldwide. Every heartbeat could be your last, making heart attacks a primary concern. While some risk factors for heart disease are inherited, many can be prevented. Your daily choices, such as your diet, exercise, and stress management, impact your heart health. By making minor changes to your routine, you can reduce your risk of heart attacks and improve your overall health. Here are five important habits that can help you and may even save your life.

    What are 5 ways you can prevent and reduce the risks of a heart attack?(Adobe Stock)

    What is a balanced diet for the heart?

    What you put on your plate matters more than you think. A diet loaded with saturated fats, processed foods, and excess sugar can dramatically elevate your cholesterol levels, trigger weight gain, and skyrocket your blood pressure, all key culprits contributing to heart disease, as per the Centers for Disease Control and Prevention.

    Instead of falling into these dietary traps, focus on embracing a heart-friendly diet:

    1. Fruits and vegetables: “Bursting with antioxidants, fibre, and important vitamins, these powerhouses fortify your heart”, Dr Gautam Rege, Interventional Cardiologist, Jupiter Hospitals, tells Health Shots.
    2. Whole grains, such as oats, brown rice, and whole wheat bread, can effectively lower cholesterol levels according to The Nutrition Source.
    3. Lean proteins: Opt for fish, beans, lentils, and skinless poultry to boost your protein intake without the unhealthy fats, according to the UK’s National Health Institute.
    4. Good fats: Incorporate heart-protective unsaturated fats from sources like olive oil, nuts, seeds, and avocados into your meals as per Harvard Health.
    5. Pro Tip: Follow the “80-20 rule”, aim for 80% of your diet to be made up of nutrient-dense whole foods while allowing for 20% to enjoy occasional indulgences.

    How to stay physically active?

    Your heart is a muscle, and like any muscle, it thrives on regular exercise, as per the National Heart, Lung, and Blood Institute. A sedentary lifestyle invites a host of cardiovascular issues. In contrast, physical activity strengthens your heart, enhances blood circulation, reduces cholesterol, and maintains a healthy weight, all of which are critical factors in lowering heart attack risks.

    1. Aim for 30 minutes daily: Aim for at least 30 minutes of moderate activity, such as brisk walking, cycling, or swimming, five times a week.
    2. Include strength training: “Engage in strength training exercises two to three times per week to build lean muscle mass, which aids in regulating blood sugar and metabolism”, shares the cardiologist.
    3. Small changes count: Even simple adjustments, like opting for stairs over elevators or taking a walking break after meals, can accumulate significant health benefits.
    4. Consistency is key: Focus on enjoyable activities that encourage a daily habit of movement as per the British Heart Foundation.

    How to manage stress to reduce heart attack risk?

    Chronic stress is a silent killer, unleashing harmful hormones like cortisol and adrenaline that can elevate blood pressure and overstrain your heart. Moreover, stress often drives unhealthy coping mechanisms such as overeating, smoking, or increased alcohol consumption, as per Harvard Health.

    To protect your heart against the attack of stress, consider these methods:

    1. Mindful breathing or meditation: Dedicate 10–15 minutes daily to these practices to reduce stress levels, as per the American Heart Association.
    2. Engage in relaxing hobbies: “Immerse yourself in enjoyable activities like gardening, painting, or reading that can divert your mind from stressors”, explains Dr Rege.
    3. Prioritise sleep: Aim for quality rest, ideally 7–8 hours per night, as poor sleep can escalate stress and adversely affect your heart health, as per the American Heart Association.
    4. Stay socially connected: Engaging with family or friends can significantly lighten emotional burdens, as per Innovation in Aging.

    How to detox your body from smoking and alcohol?

    Smoking isn’t just bad; it’s downright lethal for your heart. It wreaks havoc on blood vessels, diminishes oxygen levels in the blood, and accelerates plaque buildup in arteries, significantly elevating heart attack risk as per the National Heart, Lung, and Blood Institute.

    • Kick the habit: Quitting smoking can drastically reduce your heart attack risk within months. If you find it challenging, professional help, support groups, or nicotine replacement therapies can ease the process, as per the American Cancer Society.
    • Limit alcohol consumption: Excessive alcohol intake raises blood pressure and can trigger heart rhythm problems. If you choose to drink, moderation is essential, with no more than one drink per day for women and two for men, as recommended by the Centers for Disease Control and Prevention.

    Dr Gautam Rege says, “the safest option for your heart is to avoid alcohol entirely”

    How to monitor your health daily?

    Prevention paired with awareness is key. Many heart risk factors, including high blood pressure, high cholesterol, and diabetes, often whisper silently until it’s too late, as per the Centers for Disease Control and Prevention. Regular health check-ups can help you catch problems early on.

    1. Annual check-ups: Schedule to have your blood pressure, cholesterol, and blood glucose checked at least once a year as per the American Heart Association.
    2. Track your weight and waistline: “Abdominal fat is closely correlated with increased heart disease risk; keeping an eye on these metrics can provide valuable insights”, explains the cardiologist.
    3. Stay alert for symptoms: Consult a doctor immediately if you experience warning signs like chest pain, shortness of breath, or abnormal fatigue.

    Continue Reading

  • Safety at Speed: How Maersk Delivered High-Risk EV Batteries in 72 Hours, Fully Compliant and Incident-Free

    Safety at Speed: How Maersk Delivered High-Risk EV Batteries in 72 Hours, Fully Compliant and Incident-Free

    Maersk Air Freight enabled a global EV leader to meet critical production deadlines through precision dangerous goods logistics.


    The Customer

    A leading electric vehicle (EV) manufacturer required urgent air freight for lithium-ion batteries, classified as UN3480, Class 9 Dangerous Goods, from China to Latin America. These high-energy batteries are crucial to the customer’s EV assembly operations, and shipping them by air presents significant safety and regulatory challenges.

    Faced with a narrow production window and stringent transport protocols, the customer needed a logistics partner with the ability to act fast, navigate complex international regulations, and ensure 100% safety along the way. That’s where Maersk Air Freight stepped in.

    The Challenge

    The transportation of lithium-ion batteries presents unique complexities that challenge even experienced logistics providers. These UN3480 Class 9 dangerous goods carry inherent risks, including thermal runaway, potential short circuits, and fire hazards that require specialized handling protocols.

    The customer faced three critical obstacles. First, navigating the intricate web of IATA dangerous goods regulations while ensuring full compliance across multiple international jurisdictions.

    Second, managing the safety risks associated with lithium battery transport, where even minor documentation errors could result in shipment rejection or safety incidents.

    Third, meeting urgent delivery deadlines is essential for maintaining production line continuity; any delays would cascade through their entire manufacturing schedule, potentially costing millions in lost production.

    Standard air freight solutions were inadequate for this high-stakes scenario, requiring a logistics partner with certified expertise in handling dangerous goods and the operational flexibility to deliver within a 72-hour window.

    The Solution

    Leveraging its position as an IATA-certified dangerous goods operator, Maersk Air Freight implemented a comprehensive solution tailored to the customer’s specific requirements:

    • Own Controlled Flight Network: Maersk utilized its controlled flight network, which streamlined transportation touchpoints and eliminated the complexity of multiple carrier handoffs. This controlled network proved particularly valuable when Maersk expanded its China-US route to include Latin America in May, directly aligning with the customer’s delivery destinations.
    • Pre-Shipment Compliance: Maersk’s dangerous goods specialists ensured all batteries were charged to precisely ≤ 30% State of Charge (SoC), the maximum safe level for air transport, meeting strict IATA regulations for lithium battery transportation.
    • Secure Packaging & Labeling: The team implemented rigorous packaging protocols, applying Class 9 hazard labels, CAO (Cargo Aircraft Only) markings, and specialized Lithium Battery Handling stickers to guarantee full regulatory compliance across all international jurisdictions.
    • Temperature-Controlled Logistics: Maersk’s specialists continuously monitored storage and transit temperatures throughout the journey, preventing thermal exposure that could trigger hazardous battery reactions and ensuring product integrity.
    • Expert Documentation Management: Maersk’s IATA-certified specialists handled all dangerous goods documentation, eliminating the risk of regulatory rejections and ensuring seamless customs clearance at each transit point.

    The Result

    Within just 72 hours, Maersk Air Freight delivered the Class 9 lithium-ion battery shipment from China to Latin America on time, fully compliant, and with zero incidents, rejections, and delays throughout the entire transport process.

    The cargo moved seamlessly through our controlled flight network, with all safety protocols, documentation, and temperature conditions met across every leg of the journey.

    This execution not only ensured uninterrupted EV production for the customer but also eliminated risk points such as customs rejection, delays, or mishandling during transfer.

    What began as a high-risk, high-pressure movement has now evolved into a model for repeatable success. The customer has since expanded their partnership with Maersk to cover additional dangerous good lanes in Asia and North America, confident in our ability to balance regulatory precision with operational speed. For mission-critical cargo, such as lithium batteries, Maersk has proven to be more than just a logistics provider; we’re a trusted safeguard for performance, safety, and supply chain continuity.

    Take the Next Step with Maersk

    In industries where a single delay can ripple across markets, hazardous cargo must be transported with zero error. Maersk Air Freight offers not just capacity, but certified precision, regulatory confidence, and unmatched control.

    Discover our Air Freight Solutions or learn how we support logistics for dangerous goods at scale.

    Continue Reading

  • Jaguar Land Rover extends production shutdown after cyber-attack | Jaguar Land Rover

    Jaguar Land Rover extends production shutdown after cyber-attack | Jaguar Land Rover

    Jaguar Land Rover has extended its shutdown on car production, as Britain’s biggest carmaker grapples with the aftermath of a cyber-attack.

    JLR said on Tuesday it will freeze production until at least next Wednesday, 24 September, as it continues its investigations into the hack, which first emerged earlier this month.

    The manufacturer said: “We have taken this decision as our forensic investigation of the cyber incident continues, and as we consider the different stages of the controlled restart of our global operations, which will take time.

    “We are very sorry for the continued disruption this incident is causing and we will continue to update as the investigation progresses.”

    JLR, which is owned by India’s Tata group, stopped production at its sites after discovering hackers had infiltrated its systems a few weeks ago.

    The company has since found the attack has affected “some data”, although it said that it could not provide more details of which data was affected, or if customers’ or suppliers’ information was stolen, but that it would contact anyone affected.

    The costs of the cyber-attack are likely building for JLR, as production at its factories in the Midlands and Merseyside are put on hold. Other production facilities around the world have also been affected, fuelling speculation that it could be weeks until systems are operational.

    The freeze has also affected suppliers and retailers for JLR, with some operating without computer systems and databases normally used for sourcing spare parts for garages or registering vehicles.

    skip past newsletter promotion

    Last week, the Unite union warned that thousands of workers in the JLR supply chain are at risk of losing their livelihoods, and urged the government to step in with a furlough scheme to support them.

    A group of hackers, linked to other serious hacks this year on retailers including M&S, have claimed responsibility for the attack on JLR. Screenshots which are allegedly of JLR’s internal IT systems were posted on a Telegram channel that combined the names of groups of hackers known as Scattered Spider, Lapsus$ and ShinyHunters.

    Continue Reading