Thursday, November 20, 2025

Healthcare Teams: Understanding Mask-Wearing Attitudes During COVID-19 #pencis #researchawards

 


Introduction

The emergence of COVID-19 heightened global reliance on protective equipment, particularly face masks, making them a central public health measure. This study investigates how socio-demographic characteristics—such as education level and occupation—shape public perception of interprofessional healthcare teams wearing FFP2 masks. Through an online survey of 906 participants assessing attitudes toward team photographs across various attire scenarios, the research aims to uncover implicit beliefs regarding mask effectiveness. By integrating elements of the Health Belief Model (HBM), the study provides insight into how individual attitudes toward preventive behaviors influence real-world decision-making during pandemics.

Influence of Socio-Demographic Factors on Mask Perception

Socio-demographic factors play a critical role in shaping public attitudes toward preventive health measures such as mask-wearing. The findings show that participants’ education and occupational background significantly influenced how they perceived interprofessional teams wearing FFP2 masks. These variations highlight the complexity of public health communication, where different population groups may interpret the same protective behavior through diverse personal, cultural, and professional lenses. Understanding these differences is essential for designing more inclusive and effective health policies.

Perception Differences Between Healthcare Workers and the General Public

A notable finding of the study is the contrasting attitude between healthcare professionals and individuals from other occupational sectors. Healthcare workers appeared more critical of interprofessional teams wearing FFP2 masks, with nearly one-third expressing doubts about mask effectiveness. This skepticism underscores an intriguing paradox: those with the highest exposure to medical knowledge and clinical environments may also develop unique biases shaped by situational fatigue, professional experience, and risk perception. Exploring this contrast is vital in addressing gaps in compliance and communication strategies.

Role of Implicit Attitudes in Public Health Behavior

By examining perceptions of mask-wearing indirectly through the evaluation of team photographs, the study captures implicit attitudes that may not always align with participants’ openly stated beliefs. These underlying perceptions are crucial in shaping real-life behaviors, particularly when individuals face ambiguous or evolving health guidelines. Implicit attitudes can subtly influence adherence to preventive measures, making it important for public health interventions to acknowledge and address both conscious and unconscious factors driving behavior.

Application of the Health Belief Model (HBM) in Understanding Mask Attitudes

The Health Belief Model provides a theoretical foundation to interpret how perceptions of susceptibility, severity, benefits, and barriers influence adherence to mask-wearing. The study successfully uses HBM to connect participants’ perceptions of interprofessional teams wearing FFP2 masks with broader beliefs about COVID-19 protection. Healthcare professionals’ skepticism, for instance, may reflect perceived barriers or low perceived benefits. Applying HBM offers a deeper understanding of how cognitive processes guide preventive behaviors during a health crisis.

Implications for Future Pandemic Preparedness

The results emphasize the importance of addressing perception gaps among both healthcare workers and the general population to strengthen preparedness for future pandemics. Recognizing the diversity of attitudes toward basic protective measures like mask-wearing can guide the development of targeted educational campaigns, optimized communication strategies, and improved trust-building within communities. These insights are crucial not only for COVID-19 but also for designing adaptable frameworks for future infectious disease outbreaks.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/
📧 Contact us: infectioussupport@pencis.com

Hashtags

#COVID19Research, #MaskPerception, #FFP2Masks, #HealthcareTeams, #HealthBeliefModel, #PandemicPreparedness, #PublicHealthAttitudes, #SociodemographicFactors, #HealthcareProfessionals, #PreventiveMeasures, #BehavioralHealth, #ImplicitAttitudes, #HealthCommunication, #COVID19Prevention, #InterprofessionalCare, #MedicalResearch, #PublicHealthPolicy, #EpidemiologyStudy, #InfectionControl, #HealthSurvey,

Monday, November 17, 2025

Optimization of Postoperative Antimicrobial Therapy Using CDSS


Introduction

Optimizing postoperative antimicrobial therapy is a major priority in modern surgical practice due to rising antimicrobial resistance and the persistent risk of healthcare-associated infections. The integration of Clinical Decision Support Systems (CDSSs) has emerged as an effective strategy to guide evidence-based prescribing, standardize antimicrobial use, and enhance patient safety. In the context of a multi-year quasi-experimental study, CDSS implementation in a primary-level hospital demonstrated measurable improvements in antimicrobial stewardship within diverse surgical specialties. By analyzing antimicrobial consumption, expenditure trends, and clinical outcomes such as mortality and length of stay, this research highlights the transformative potential of informatics-driven interventions in surgical environments. The findings underscore how targeted digital stewardship can support more rational drug use without compromising patient outcomes.

Impact of CDSS on Antimicrobial Consumption in Surgical Patients

Clinical Decision Support Systems significantly influenced postoperative antimicrobial prescribing patterns by promoting more rational and standardized usage. A 4.4% overall reduction in antimicrobial consumption demonstrates the efficiency of CDSS-driven optimization, while large decreases in specific drug classes such as aminoglycosides (−52.0%), macrolides, lincosamides and streptogramins (−40.6%), and fluoroquinolones (−32.3%) indicate targeted improvements. These reductions reflect a shift away from broad-spectrum or high-toxicity agents, supporting safer and more appropriate therapeutic choices. This research reveals the value of CDSS tools in modifying prescribing behavior and reducing unnecessary exposure to antimicrobials, which in turn helps limit the development of antimicrobial resistance.

Variability in Antimicrobial Stewardship Outcomes Across Surgical Specialties

The study illustrates how the impact of CDSS may differ across surgical services due to variations in workflow, infection risk, and prescribing habits. Traumatology and Urology showed the most significant reductions in antimicrobial consumption—21.3% and 14.3%, respectively—highlighting areas where CDSS recommendations aligned strongly with clinical practice. These heterogeneous outcomes reinforce the importance of specialty-specific stewardship strategies and suggest that tailored CDSS algorithms may further enhance prescribing accuracy. Understanding the dynamics within individual surgical departments enables hospitals to refine stewardship interventions for maximum clinical and operational benefit.

Influence of CDSS on Antimicrobial Expenditures and Hospital Resource Optimization

The downward trend in antimicrobial expenditures—from 3185.4 to 2733.9 €/1000 patient-days (−14.2%)—demonstrates that CDSS-supported stewardship can effectively reduce pharmacy costs. Although the change did not reach statistical significance (p = 0.17), the financial impact indicates potential long-term savings, especially in high-consumption settings. Reduced use of expensive broad-spectrum antibiotics directly contributes to budget optimization, while improved prescribing efficiency may also reduce downstream costs associated with adverse events or antimicrobial resistance. This topic highlights the economic value of integrating CDSS technologies within hospital stewardship programs.

Clinical Outcomes: Mortality Stability and Reductions in Length of Stay

A central strength of the CDSS-driven antimicrobial stewardship approach is that it improved resource utilization without compromising patient safety. Mortality rates remained stable, confirming that reduced antimicrobial exposure did not negatively affect clinical outcomes. Significant reductions in length of stay (LOS) were observed in key surgical services—Urology (5 to 4 days) and Traumatology (16 to 8.5 days)—suggesting faster recovery and fewer complications. These improvements reflect timely de-escalation, discontinuation, and optimization of therapy, supporting the role of CDSS in enhancing both patient flow and clinical efficiency within surgical departments.

Stewardship Interventions and Acceptance of CDSS Recommendations

High acceptance of stewardship recommendations (76.1%) demonstrates strong clinician engagement with CDSS-guided antimicrobial management. The most frequent interventions—discontinuation of antimicrobials (25.8%), transition to oral therapy (21.0%), and de-escalation (18.7%)—reflect core stewardship principles aimed at minimizing unnecessary exposure while maintaining therapeutic effectiveness. The substantial number of recommendations issued (476 for 330 patients) highlights the proactive nature of the CDSS in identifying optimization opportunities. This topic emphasizes how integrating decision support into clinical workflows can strengthen stewardship culture, enhance prescriber confidence, and promote sustainable antimicrobial use.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/

📧 Contact us: infectioussupport@pencis.com

Hashtags

#AntimicrobialStewardship, #SurgicalInfections, #ClinicalDecisionSupport, #PostoperativeCare, #HospitalEpidemiology, #AntimicrobialResistance, #CDSSImpact, #SurgicalOutcomes, #DrugUtilization, #HealthInformatics, #StewardshipInterventions, #PatientSafety, #LengthOfStay, #AntibioticOptimization, #HealthcareQuality, #AntibioticConsumption, #EvidenceBasedMedicine, #DigitalHealthTools, #Pharmacoeconomics, #AntibioticExpenditures,

Saturday, November 15, 2025

MRI Reflects Meningioma Biology and Molecular Risk | Advanced Tumor Imaging #pencis #researchawards


Introduction

Meningiomas represent the most common primary intracranial tumors, and recent advances in large-scale genomic and epigenomic profiling have reshaped their biological classification frameworks. The cIMPACT-NOW update 8 has highlighted the importance of integrating molecular signatures—such as DNA methylation classes and chromosomal alterations—into clinical decision-making. As precision oncology grows, one key question is whether non-invasive imaging can reflect these complex molecular landscapes. Magnetic Resonance Imaging (MRI), with its rich structural and textural information, offers a promising avenue. By incorporating radiomics and machine-learning models, researchers aim to capture molecular heterogeneity directly from imaging, potentially reducing dependency on invasive tissue sampling. This introduction sets the foundation for exploring how radiomics-based MRI analysis may transform risk stratification and treatment planning in meningioma management.

Advances in Molecular Profiling of Meningiomas

Large-scale epigenomic studies have uncovered significant molecular diversity within meningiomas, moving beyond conventional WHO grading. DNA methylation profiling and copy-number variation analyses have identified biologically distinct tumor subgroups with different prognoses and therapeutic implications. These molecular systems provide a more accurate prediction of recurrence risk and treatment response compared to histopathology alone. As molecular criteria increasingly supplement or supersede traditional grading, understanding how these patterns correlate with imaging characteristics becomes essential. The integration of these datasets underscores the evolution toward biologically driven classification frameworks.

Radiomics: A Non-Invasive Window Into Tumor Biology

Radiomics has emerged as a powerful tool that transforms MRI scans into quantitative datasets capturing tissue heterogeneity, morphology, and microenvironment features. By leveraging advanced segmentation frameworks such as BraTS pipelines, researchers extract high-dimensional features from tumor core and edema regions. These features can correlate with underlying molecular alterations, enabling models to infer biological risk signatures. Radiomics thereby offers a non-invasive “imaging biomarker” approach, potentially allowing earlier and more accessible prediction of aggressive behavior or specific genetic alterations before surgical sampling.

Machine Learning Models for Molecular Risk Prediction

Random Forest classifiers trained on radiomic signatures have shown strong diagnostic potential in predicting molecular risk categories within meningiomas. In particular, accuracy above 91% for integrated molecular risk groups demonstrates that radiomic patterns align closely with epigenetic and copy-number alterations. The ability to predict specific markers such as 1p loss with 87.5% accuracy and AUC 0.90 further supports the biological sensitivity of radiomics. These findings highlight how machine learning models can bridge imaging and molecular diagnostics, providing rapid, automated assessments that may guide preoperative planning.

Challenges in Predicting WHO Grade Using MRI

Despite promising results in molecular risk prediction, radiomics-based models show inferior performance in predicting WHO grade, with accuracy around 76.8%. This limitation emphasizes the imperfect alignment between histopathological grading criteria and MRI-based phenotypes. Tumor grade often relies on microscopic features such as mitotic count and necrotic foci that are not always reflected in gross radiographic appearance. This discrepancy suggests that while MRI captures molecular risk effectively, WHO grade remains more challenging due to its histology-centric foundation. Future studies should explore integrating radiomics with advanced MRI sequences or multimodal imaging to improve prediction accuracy.

Clinical Implications and Future Research Directions

The ability of MRI-radiomics models to approximate key molecular attributes offers significant clinical potential. Preoperative prediction of high-risk molecular signatures could help prioritize surgical strategies, biopsy planning, early referral for adjuvant therapy, or expedited confirmatory molecular testing. However, transitioning these models into routine clinical pathways requires robust external validation, harmonization across MRI scanners, and integration with prospective trials. Future research should broaden datasets across institutions, enhance model interpretability, and explore multimodal approaches combining radiomics, genomics, and liquid biopsy data. Ultimately, these advancements may redefine meningioma management through precision imaging.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/
📧 Contact us: infectioussupport@pencis.com

Hashtags

#MeningiomaResearch, #Radiomics, #MRIImaging, #MolecularDiagnostics, #Epigenomics, #cIMPACTNOW, #BrainTumorBiology, #MachineLearningInMedicine, #AIInRadiology, #NeuroOncology, #CopyNumberVariation, #DNA_Methylation, #TumorClassification, #PrecisionMedicine, #MedicalImagingAI, #Radiogenomics, #WHOGrade, #ClinicalDecisionSupport, #NeuralTumors, #BiomedicalResearch,

Friday, November 14, 2025

High-Throughput Discovery of Monkeypox H1 Phosphatase Inhibitors #pencis #researchawards

Introduction

Mpox, caused by the monkeypox virus (MPXV), continues to pose a profound global health concern as it remains classified as a Public Health Emergency of International Concern (PHEIC). Despite its expanding burden, therapeutic strategies targeting MPXV remain limited, underscoring the urgent need for novel antiviral approaches. Recent research has highlighted the orthopoxvirus dual-specificity phosphatase H1 as a critical regulator of MPXV pathogenesis, making it an attractive antiviral target. H1 contributes to viral immune evasion, early transcriptional regulation, and viral particle maturation, and its suppression significantly decreases viral infectivity. These insights drive scientific efforts to explore H1 inhibition as a pathway toward next-generation therapeutic development. This study advances such efforts by establishing a high-throughput platform to identify potent small-molecule inhibitors of MPXV H1.

H1 Phosphatase as a Therapeutic Target

The H1 phosphatase is a multifunctional viral enzyme that plays a central role in MPXV immune evasion and replication, making it a prime candidate for targeted antiviral intervention. By suppressing interferon signaling and regulating critical stages of viral transcription and core protease activity, H1 ensures efficient viral propagation within host cells. Research reveals that downregulating H1 dramatically reduces the number of infectious viral particles produced, confirming the enzyme's indispensability in MPXV's life cycle. These findings identify H1 not just as a structural component, but as a cornerstone of viral pathogenicity, making it one of the most promising molecular targets for therapeutic innovation.

High-Throughput Screening Assay Development

To accelerate the discovery of MPXV H1 inhibitors, a robust and efficient high-throughput enzymatic assay was developed using p-nitrophenyl phosphate (pNPP) as a substrate. This assay allows rapid quantification of H1 phosphatase activity and provides a scalable platform for screening large chemical libraries. Key structural elements—including the N-terminal helix α1 and the catalytic residue Cys110—were validated as essential contributors to enzyme functionality, reinforcing their importance in inhibitor design. The assay’s sensitivity and reproducibility make it suitable for identifying potent compounds capable of disrupting H1 function.

Structural Hot Spots: Helix α1 and Cys110

Detailed functional analyses confirmed that both helix α1, which mediates H1 dimerization, and the catalytic cysteine residue Cys110 are indispensable for enzymatic activity. Mutational studies demonstrated that disrupting either structural region leads to significant loss of enzymatic function, emphasizing their relevance as strategic “hot spots” for rational antiviral design. These structural determinants not only govern catalytic efficiency but also influence substrate accessibility and protein stability. This knowledge enhances structure-guided drug design efforts aimed at generating inhibitors that can effectively disrupt key interactions or catalytic mechanisms within H1.

Discovery of Potent MPXV H1 Inhibitors

Using the high-throughput assay, researchers identified 17 potent small molecules with nanomolar IC50 values and minimal cytotoxicity. These inhibitors demonstrated robust suppression of H1 enzymatic activity, highlighting their potential as lead compounds for further antiviral development. The discovery of molecules that exhibit both potency and safety underscores the promise of targeting H1 as a therapeutic strategy. Moreover, the chemical diversity of these inhibitors expands the landscape of potential antiviral scaffolds that can be further optimized through medicinal chemistry approaches.

Molecular Docking and Mechanistic Insights

Computational molecular docking studies revealed that the newly discovered inhibitors bind tightly within the active site of MPXV H1, interacting with residues in the P-loop and WPD-loop. These interactions effectively restrict substrate entry, thereby suppressing phosphatase activity. The structural alignment of inhibitor binding modes offers valuable insights into the molecular mechanisms underlying H1 inhibition and provides a blueprint for structure-based drug optimization. These findings collectively strengthen the rationale for advancing H1-targeted antivirals into preclinical development pipelines.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/
📧 Contact us: infectioussupport@pencis.com

Hashtags

#MpoxResearch, #MonkeypoxVirus, #MPXV, #H1Phosphatase, #AntiviralDiscovery, #DrugDevelopment, #HighThroughputScreening, #ViralPathogenesis, #Orthopoxvirus, #MolecularDocking, #InhibitorDesign, #GlobalHealthThreat, #InterferonSignaling, #StructuralBiology, #EnzymaticAssay, #NanomolarInhibitors, #TherapeuticTargets, #VirologyResearch, #EmergingViruses, #ScienceFatherResearchAwards,

Thursday, November 13, 2025

Childhood Infectious Diseases: Understanding Causes,Prevention,Global Impact #pencis #researchawards


Introduction

Infectious diseases remain a leading cause of child mortality worldwide, claiming millions of young lives annually. Among the estimated 10 million deaths of children under five each year, a substantial proportion results from preventable infections such as pneumonia, diarrhea, malaria, measles, and neonatal sepsis. Despite advancements in global health, the burden remains disproportionately high in low- and middle-income countries, where healthcare access and preventive interventions are limited. Evidence-based strategies, including vaccination, improved hygiene, breastfeeding promotion, and timely treatment, can significantly reduce this mortality rate. Addressing these issues through research, policy, and implementation remains an urgent global health priority.

The Global Burden of Infectious Diseases in Children

Child mortality due to infectious diseases highlights stark inequalities in global health outcomes. About 42 countries account for nearly 90% of global under-five deaths, emphasizing the need for targeted interventions. Neonatal infections, particularly sepsis, pneumonia, diarrhea, and tetanus, remain the most common causes of early childhood deaths. Understanding regional disparities, socio-economic determinants, and healthcare delivery challenges is critical for designing effective prevention and treatment strategies to combat this persistent global health issue.

Preventable Causes and Public Health Strategies

A significant portion of childhood deaths from infectious diseases is preventable through low-cost, evidence-based interventions. Strategies such as maternal immunization, clean delivery practices, breastfeeding, improved sanitation, and vaccination programs can drastically reduce mortality. Research focused on the implementation and scalability of these interventions can enhance their effectiveness, ensuring that life-saving measures reach the most vulnerable populations.

 Advances in Neonatal Infection Prevention

Preventing newborn infections requires a multifaceted approach that begins with maternal health. Tetanus toxoid vaccination, sterile birth environments, and proper umbilical cord care are crucial preventive measures. Furthermore, promoting exclusive breastfeeding and early detection of infections significantly improve neonatal survival rates. Ongoing research on neonatal immune responses and antimicrobial resistance patterns provides valuable insights into improving infection control in early life.

Diarrheal and Respiratory Disease Control

Diarrhea and pneumonia are responsible for nearly half of infectious disease deaths in children under five. Interventions such as oral rehydration therapy, zinc supplementation, rotavirus and pneumococcal vaccination, and improved nutrition have proven to reduce mortality. Continued research on microbial pathogenesis, environmental hygiene, and vaccine development remains essential to sustain and enhance these gains in child health outcomes.

Scaling Up Interventions for Sustainable Impact

While effective interventions exist, their global implementation remains a challenge. Scaling up requires integrated efforts involving policy reform, community engagement, and resource mobilization. Research should focus on health systems strengthening, behavioral change models, and cost-effective delivery methods to ensure sustained impact. The ultimate goal is to build resilient healthcare systems capable of preventing and managing infectious diseases in children globally.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/
📧 Contact us: infectioussupport@pencis.com

Hashtags

#ChildHealthResearch, #InfectiousDiseases, #PneumoniaPrevention, #DiarrhealDiseases, #NeonatalSepsis, #GlobalChildMortality, #MaternalHealth, #VaccineResearch, #PublicHealthInterventions, #ZincTherapy, #RotavirusVaccine, #HygienePromotion, #CleanDelivery, #OralRehydration, #HealthSystemsResearch, #PediatricInfections, #ScienceFather, #ResearcherAwards, #ChildSurvival, #GlobalHealth,

Wednesday, November 12, 2025

Genomics Network | Advancing Pathogen Genomics for Public Health in Australia#pencis #researchawards



 Introduction

The COVID-19 pandemic underscored the transformative potential of pathogen genomics in enhancing global public health surveillance and response. Genomic technologies allow rapid identification, tracking, and understanding of infectious agents, enabling more informed and timely interventions. In Australia, this realization led to the establishment of the Communicable Diseases Genomics Network (CDGN) in 2015. The CDGN serves as a bridge between laboratories, researchers, and policymakers, ensuring genomic data are efficiently utilized to improve disease control strategies. Its success has positioned Australia as a global model for integrating genomics into public health systems.

The Role of CDGN in Coordinating Pathogen Genomics

The CDGN plays a critical role in harmonizing national efforts to integrate pathogen genomics within public health systems. By establishing standardized frameworks for genomic data sharing, ethical governance, and laboratory coordination, CDGN promotes consistency and interoperability across jurisdictions. Its coordinated model ensures rapid, accurate, and transparent genomic surveillance during public health emergencies. The network’s unified approach has strengthened communication between public health laboratories, fostering collaboration that accelerates research translation and response readiness.

Translational Research and Policy Development

One of the defining achievements of CDGN is its contribution to translational research and evidence-based policymaking. By connecting genomic insights with real-world public health challenges, CDGN ensures that data-driven policies are informed by robust scientific evidence. This approach supports the creation of responsive health frameworks capable of addressing both emerging and re-emerging infectious threats. The outcomes of CDGN-supported research have guided national decision-making on outbreak control, vaccine strategies, and genomic data regulation, ultimately enhancing public trust in genomic science.

Workforce Capacity Building and Training

Effective implementation of pathogen genomics requires a skilled workforce capable of managing, analyzing, and interpreting genomic data. CDGN has prioritized capacity building through training initiatives, workshops, and knowledge-sharing programs that empower public health professionals and researchers. These programs enhance bioinformatics expertise, laboratory skills, and interdisciplinary collaboration. By investing in human capital, CDGN not only strengthens Australia’s domestic public health infrastructure but also contributes to the global genomic surveillance ecosystem.

Data Sharing, Governance, and Ethical Frameworks

Ethical governance and secure data sharing are foundational to the success of pathogen genomics. CDGN has established standardized governance models that balance transparency, privacy, and collaboration. Its frameworks ensure responsible use of genomic data, promoting trust between institutions, governments, and the public. The network’s emphasis on open data principles while maintaining stringent ethical standards has set an international benchmark for federated genomic systems and has encouraged global alignment toward responsible data stewardship.

Global Implications and Future Directions

The success of the Communicable Diseases Genomics Network demonstrates how national collaboration can translate into global leadership in public health genomics. CDGN’s model, grounded in cross-sector coordination and innovation, provides a scalable blueprint for other federated nations. Future directions include expanding real-time genomic surveillance, integrating artificial intelligence for predictive analytics, and strengthening international genomic data exchange. Through these initiatives, the CDGN continues to drive advancements that safeguard populations and inform global health preparedness.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/
📧 Contact us: infectioussupport@pencis.com

Hashtags 

#PathogenGenomics, #PublicHealthInnovation, #CDGN, #GenomicSurveillance, #CommunicableDiseases, #COVID19Lessons, #GenomicResearch, #PublicHealthAustralia, #DataSharing, #HealthGovernance, #TranslationalResearch, #Bioinformatics, #HealthPolicy, #Epidemiology, #GlobalHealthSecurity, #OutbreakResponse, #HealthTechnology, #WorkforceDevelopment, #PrecisionPublicHealth, #ScienceFatherResearchAwards,

Tuesday, September 23, 2025

Betaine & Lung Health 🫁 | Pulmonary Macrophage Pyroptosis Inhibition | #pencis #FOXO1 #LungInjury



INTRODUCTION

Bronchopulmonary dysplasia (BPD) remains one of the most prevalent chronic lung diseases in premature infants, characterized by impaired alveolar development and long-term respiratory complications. Emerging research highlights the importance of inflammatory pathways, particularly NLRP3-mediated macrophage pyroptosis, in the pathogenesis of BPD. Pyroptosis, a pro-inflammatory form of cell death, contributes to excessive inflammation and tissue injury in the immature lung exposed to hyperoxia. In this context, betaine, a naturally occurring compound with well-established anti-inflammatory and antioxidant properties, has attracted scientific interest as a potential therapeutic candidate. By modulating molecular signaling pathways, including FOXO1 phosphorylation, betaine may offer protective effects against hyperoxia-induced lung injury and provide new insights into treatment strategies for BPD.

PATHOGENESIS OF BPD AND NLRP3-MEDIATED PYROPTOSIS

BPD pathogenesis is multifactorial, involving mechanical ventilation, oxygen toxicity, and inflammatory responses that disrupt normal lung development. Among these, macrophage-driven inflammation through NLRP3 inflammasome activation plays a central role. Hyperoxia significantly increases the expression of pyroptosis-associated proteins, leading to alveolar simplification and impaired vascular growth. Pyroptotic macrophages release inflammatory cytokines, which exacerbate pulmonary damage and hinder alveolarization. Therefore, targeting NLRP3-mediated macrophage pyroptosis has emerged as a promising therapeutic strategy to improve outcomes in preterm infants with BPD.

ROLE OF BETAINE IN ANTI-INFLAMMATORY MODULATION

Betaine acts as a methyl donor in metabolic processes and exerts strong anti-inflammatory and antioxidative functions. In the context of BPD, betaine reduces oxidative stress markers and inflammatory mediators while preserving lung tissue integrity. Experimental evidence demonstrates that daily subcutaneous administration of betaine in neonatal mice exposed to hyperoxia significantly reduces macrophage pyroptosis. By attenuating oxidative injury and inflammatory cytokine production, betaine supports both structural and functional lung protection, indicating its therapeutic potential.

FOXO1 PHOSPHORYLATION AND BETAINE INTERVENTION

The transcription factor FOXO1 is closely associated with cell survival, inflammation, and oxidative stress responses. Hyperoxia induces FOXO1 phosphorylation, which in turn promotes NLRP3 activation and pyroptosis in pulmonary macrophages. Betaine has been shown to inhibit the phosphorylation of FOXO1, thereby preventing NLRP3 activation and subsequent pyroptotic cell death. In vitro studies using RAW264.7 macrophages confirmed that betaine suppressed FOXO1 phosphorylation and pyroptosis under hyperoxic conditions, while treatment with okadaic acid, a phosphatase inhibitor, reversed these protective effects.

IMPACT ON LUNG DEVELOPMENT

Hyperoxia-induced injury impairs alveolarization, leading to fewer and larger alveoli typical of BPD. Betaine treatment has been found to restore alveolar structure, reduce inflammatory infiltration, and promote lung development in neonatal mice exposed to hyperoxia. By modulating molecular pathways and reducing macrophage pyroptosis, betaine indirectly supports lung growth and enhances overall pulmonary architecture. This suggests that betaine not only acts as an anti-inflammatory agent but also plays a crucial role in developmental lung protection.

FUTURE RESEARCH DIRECTIONS

The findings on betaine’s protective effects in hyperoxia-induced BPD provide a foundation for translational research. Future studies should focus on dose optimization, timing of administration, and long-term safety in neonatal populations. Investigating the interaction of betaine with other molecular pathways may also reveal synergistic therapeutic benefits. Clinical trials will be necessary to validate preclinical evidence and establish betaine as a viable adjunct therapy for preventing or treating BPD in preterm infants.

🔗 Visit: https://infectious-diseases-conferences.pencis.com
🌟 Nominate Now: https://infectious-diseases-conferences.pencis.com/award-nomination/ ecategory=Awards&rcategory=Awardee
📝 Registration page: https://infectious-diseases-conferences.pencis.com/award-registration/
📧 Contact us: infectioussupport@pencis.com

Hashtags

#BronchopulmonaryDysplasia, #BPD, #MacrophagePyroptosis, #NLRP3, #FOXO1, #Betaine, #NeonatalLungDisease, #Hyperoxia, #LungDevelopment, #PulmonaryInflammation, #Pyroptosis, #NeonatalCare, #AntiInflammatory, #OxidativeStress, #LungInjury, #MolecularTherapeutics, #PretermInfants, #RespiratoryResearch, #TranslationalMedicine, #LungHealth

Healthcare Teams: Understanding Mask-Wearing Attitudes During COVID-19 #pencis #researchawards

  Introduction The emergence of COVID-19 heightened global reliance on protective equipment, particularly face masks , making them a centr...