Preprint
Article

This version is not peer-reviewed.

Threshold Dose Response of Aluminum Adjuvants Seen in Population Data

Submitted:

18 November 2025

Posted:

18 November 2025

You are already at the latest version

Abstract

This study investigates potential associations between aluminum-adjuvanted childhood vaccines and inflammatory conditions using population-level data from the National Survey of Children's Health (NSCH) 2020-2023 and the CDC National Immunization Survey (NIS) 2011-2017. By joining datasets across 50 U.S. states and the District of Columbia for birth years 2011-2017, we analyzed vaccine uptake probabilities as proxies for aluminum exposure and prevalences of autism (3.7%), allergies (26.1%), ADHD (8.1%), asthma (8.4%), epilepsy (0.94%), obesity (4.9%), and Tourette’s syndrome (0.21%), with blood disorders (0.46%) as a negative control.Methods involved calculating disease prevalence, disease correlations, and linear regressions between vaccine likelihoods (DTaP, HepB, Hib, PCV, Polio) across age windows (0-36 months) and outcomes.Results revealed strong positive correlations among inflammatory conditions (r=0.19-0.62, p<0.001) but none with blood disorders. Aluminum exposure proxies during 6-12 months forecasted higher disease prevalence (p<0.05), with nonlinear patterns including sign reversals across time periods, consistent with NLRP3 inflammasome's two-step activation threshold.These findings suggest aluminum adjuvants may contribute to NLRP3-mediated inflammation in susceptible populations, warranting further mechanistic and prospective studies to optimize vaccination strategies and explore inflammasome-targeted therapies for reducing chronic childhood disease burdens.

Keywords: 
;  ;  

1. Introduction

According to the NSCH 2020-2023 [1,2], 41% of children have at least one of autism, allergies, adhd, asthma, epilepsy, obesity, or Tourette’s syndrome. Blood disorders (sickle cell, hemophilia, and thalassemia) are included in this study as a negative control. CDC National Immunization Survey (CDC NIS) [3] can be joined with the NSCH by US state and birth year to investigate whether vaccines have any relationship to inflammatory outcomes tracked by the NSCH. Reactogenicity from aluminum adjuvants is known to drive inflammation though its interaction with the NLRP3 inflammasome. Aluminum adjuvants are in most childhood vaccines, notably DTaP, Hib, Hep B, PCV, and Polio that were looked at in this study. NLRP3 is activated in a two step process, priming and activation, that could lead to a uniquely nonlinear dose response function for aluminum adjuvants taking the form of a threshold function. This study uses 4 years of data from the NSCH 2020-2023 joined with 7 years of data from CDC NIS to investigate the prevalence of Autism, Allergies, Asthma, ADHD, Epilepsy, and Tourette’s across the 50 states and DC from the birth years 2011-2017.

2. Methods

The study population consists of the set of people surveyed by CDC NIS 2011-2017 along with those surveyed by the NSCH 2020-2023. These two data sets were joined by birth year and US state giving 357 data points which contain exposure estimates from CDC NIS and outcome estimates from the NSCH. CDC NIS tracks vaccination uptake by age (2m, 4m, 6m, 12m, 18m, 24m, and 36m) and type of vaccine (DTap, HepB, Hib, PCV, Polio, MMR). The exposures take the form of likelihoods that the population received a particular type of vaccine in a particular age window. These likelihoods can also be summed across types to represent a proxy variable for total aluminum exposure from vaccines in a population at a particular age.
Outcomes are tracked by the NSCH, specifically these outcomes are defined by the survey questions:
  • Autism—Has a doctor or other health care provider EVER told you that this child has Autism or Autism Spectrum Disorder (ASD)? Include diagnoses of Asperger’s Disorder or Pervasive Developmental Disorder (PDD). (K2Q35A)
  • Allergies—Has a doctor or other health care provider EVER told you that this child has Allergies (including food, drug, insect, or other)?
  • Asthma—Has a doctor or other health care provider EVER told you that this child has Asthma? (K2Q40A)
  • ADHD—Has a doctor or other health care provider EVER told you that this child has Attention Deficit Disorder or Attention Deficit/Hyperactivity Disorder, that is, ADD or ADHD? (K2Q31A)
  • Blood—Has a doctor or other health care provider EVER told you that this child has Blood Disorders (such as Sickle Cell Disease, Thalassemia, or Hemophilia)?
  • Epilepsy—Has a doctor or other health care provider EVER told you that this child has Epilepsy or Seizure Disorder? (K2Q42A)
  • Obesity—Has a doctor or other health care provider ever told you that this child is overweight?
  • Tourettes—Has a doctor or other health care provider EVER told you that this child has Tourette Syndrome? (K2Q38A)
With the definitions out of the way, this study will calculate the prevalence of each disease, any associations between diseases, and finally any associations between our exposures and outcomes.

3. Results

First, the table below is the prevalence of each outcome in the NSCH.
Table 1. Prevalence of disease as measured in NSCH.
Table 1. Prevalence of disease as measured in NSCH.
Disease Prevalence
Autism 3.7%
Allergies 26.1%
ADHD 8.1%
Asthma 8.4%
Blood 0.46%
Epilepsy 0.94%
Obesity 4.9%
Tourette’s 0.21%
Every one of the inflammatory diseases is associated with each of the other inflammatory diseases. Blood disorders on the other hand have no association with the other conditions.
Table 2. Associations between disease prevalence.
Table 2. Associations between disease prevalence.
autism allergies adhd asthma blood epilepsy obesity tourettes
autism - .28*** .35*** .28*** .02 .26*** .23*** .19***
allergies .28*** - .58*** .60*** -.01 .20*** .46*** .26***
adhd .35*** .58*** - .62*** .00 .19*** .59*** .41***
asthma .28*** .60*** .62*** - .08 .22*** .52*** .32***
blood .02 -.01 .00 .08 - .02 .07 -.05
epilepsy .26*** .20*** .19*** .22*** .02 - .21*** .14**
obesity .23*** .46*** .59*** .52*** .07 .21*** - .33***
tourettes .19*** .26*** .41*** .32*** -.05 .14** .33*** -
Note. This table presents Pearson correlation coefficients. * p<.05, ** p<.01, *** p<.001.
It’s possible to forecast the prevalence of inflammatory diseases from the summed likelihood of aluminum adjuvant vaccination from CDC NIS between ages 6-12 months. Blood disorders on the other hand have no association with aluminum from vaccines.
Table 3. Future chronic childhood disease prevalence can be forecast from the summed likelihood of aluminum adjuvant vaccination from CDC NIS at ages 6-12 months.
Table 3. Future chronic childhood disease prevalence can be forecast from the summed likelihood of aluminum adjuvant vaccination from CDC NIS at ages 6-12 months.
Disease p-value t value R^2
Autism < .001 7.42 0.1173
Allergies < .001 6.63 0.0955
Asthma < .001 6.40 0.0894
ADHD < .001 6.68 0.0969
Blood 0.73 0.33 0.0003
Epilepsy .013 2.51 0.0128
Obesity < .001 5.75 0.0827
Tourettes < .001 3.60 0.0286
The summed likelihoods are shown above, but the breakdown of associations from each individual vaccine to each outcome is shown below.
Table 4. Vaccine likelihood from 6-12 months vs. chronic diseases.
Table 4. Vaccine likelihood from 6-12 months vs. chronic diseases.
DTaP HepB Hib PCV Polio
Autism .24*** .22*** .17*** .25*** .19***
Allergies .26*** .22*** .07 .28*** .17***
ADHD .30*** .15** .20*** .32*** .17***
Asthma .20*** .25*** .06 .22*** .11*
Blood -.04 .09 -.06 .00 -.03
Epilepsy .13** .05 .11* .10* .09
Obesity .19*** .33*** .03 .25*** .11*
Tourettes .09 .20** .03 .14** .03
Note. This table presents Pearson correlation coefficients. * p<.05, ** p<.01, *** p<.001.
Notably, Hib and HepB likelihoods are negatively correlated in this time period at -.12. Yet we can see that Hib and HepB are both independently associated with Autism and ADHD. That does not bode well for any hypothesis of a hidden variable. If there was a hidden variable responsible for the association, correlations between different vaccines would tend to be strictly positive from the partial information from the hidden variable.
The 4-6 month time period is interesting because of the disparate impact of the different vaccines. The meaningful difference between these vaccines is the dose of aluminum received from each one, which may be received in combination or alone with varying levels of aluminum depending on the exact brand.
Table 5. Vaccine likelihood from 4-6 months vs. chronic disease.
Table 5. Vaccine likelihood from 4-6 months vs. chronic disease.
correlation DTaP HepB Hib PCV Polio
Autism -.22*** .23*** .21*** -.26*** .21***
Allergies -.21*** .25*** .23*** -.24*** .27***
ADHD -.25*** .33*** .26*** -.29*** .27***
Asthma -.16** .25*** .20*** -.20*** .21***
Blood .08 -.03 -.01 .05 -.01
Epilepsy -.13* .12* .14* -.13* .14**
Obesity -.09 .25*** .24*** -.18*** .22***
Tourettes -.08 .15** .15** -.12* .11*
Note. This table presents Pearson correlation coefficients. * p<.05, ** p<.01, *** p<.001.
The 12-18 month time period is also interesting as once again different vaccines appear to have different relationships to future disease. Even though not every association is significant, they all have the same sign for each vaccine, once again with the exception of our negative control.
Table 6. Vaccine likelihood from 12-18 months vs. chronic disease.
Table 6. Vaccine likelihood from 12-18 months vs. chronic disease.
DTaP HepB Hib PCV Polio
Autism .07 .11* -.11* .17*** -.15**
Allergies .05 .12* -.12* .12* -.08
ADHD .10* .14** -.18*** .15** -.11*
Asthma .02 .07 -.26*** .06 -.12*
Blood -.08 -.04 .02 -.05 .07
Epilepsy .13** .10 -.07 .18*** -.02
Obesity .06 .18*** -.21*** .16** -.01
Tourettes .10 .10* -.14* .11* -.06
Note. This table presents Pearson correlation coefficients. * p<.05, ** p<.01, *** p<.001.
The table below has the summed aluminum adjuvant vaccine likelihoods for each time bucket in CDC NIS vs. each disease. This pattern of statistically significant positive and negative associations is typical of an uncorrected nonlinearity in the exposure variable.
Table 7. Aggregated vaccine likelihood vs. chronic disease for each time bucket.
Table 7. Aggregated vaccine likelihood vs. chronic disease for each time bucket.
0m-2m 2m-4m 4m-6m 6m-12m 12m-18m 18m-24m 24m-36m
Autism -.20*** -.23*** -.13* .31*** -.09 .16*** .11*
Allergies -.14** -.22*** -.07 .28*** -.08 .15** .14**
ADHD -.22*** -.27*** -.11* .31*** -.14** .20*** .10*
Asthma -.16*** -.21*** -.04 .25*** -.25*** .14** .02
Blood .04 .05 .07 .01 .01 -.02 -.12*
Epilepsy -10*. -.16** -.05 .13** -.01 .03 .09
Obesity -.06 -.15** .03 .29*** -.14** .24*** .09
Tourettes -.10 -.11* -.02 .17*** -.10 .02 .09
Note. This table presents Pearson correlation coefficients. * p<.05, ** p<.01, *** p<.001.

4. Discussion

Given that we see a non-random nonlinear associations with aluminum from vaccines at the population level, along with evidence of NLRP3 potentially mediating the nonlinear threshold dose response function of aluminum at least in asthma [4], it’s worthwhile to review all of the evidence for the role of NLRP3 inflammasome in each outcome.
NLRP3 and Autism:
The NLRP3 inflammasome, a key component of the innate immune system, plays a pivotal role in sensing cellular stress and initiating inflammatory responses through the activation of caspase-1 and the release of pro-inflammatory cytokines such as IL-1β and IL-18. Emerging evidence implicates aberrant NLRP3 activation in a spectrum of neurodevelopmental, allergic, metabolic, and neurological disorders, potentially through mechanisms involving neuroinflammation, oxidative stress, and immune dysregulation. This discussion synthesizes the current body of research linking NLRP3 to autism spectrum disorder (ASD), allergies, asthma, attention-deficit/hyperactivity disorder (ADHD), epilepsy, obesity, and Tourette’s syndrome. It further explores the two-step activation mechanism of NLRP3 and its implications for a threshold dose-response to aluminum, a known inflammasome activator. Finally, proposed treatments for each condition are examined, with emphasis on their interactions with NLRP3 pathways.
Substantial evidence supports a link between NLRP3 inflammasome activation and ASD pathogenesis, primarily through neuroinflammation and mitochondrial dysfunction. Studies have demonstrated elevated NLRP3 expression and inflammasome activity in ASD models, contributing to microglial activation, cytokine release, and behavioral deficits [5,6,7]. For instance, NLRP3-mediated IL-1β production has been associated with synaptic dysfunction and repetitive behaviors in preclinical models, with inhibition of the inflammasome rescuing immune phenotypes in vitro [8,9]. Human studies further correlate NLRP3 activity with metabolic perturbations and lipid mediators in ASD, suggesting a role in inflammasome-driven neurodevelopmental alterations [10]. These findings indicate that NLRP3 may exacerbate ASD by amplifying innate immune responses in the central nervous system (CNS), potentially triggered by environmental factors.
NLRP3 and Allergies:
NLRP3 inflammasome activation is implicated in allergic inflammation, where it promotes IL-1β and IL-18 release, driving Th2 responses and epithelial pyroptosis [11,12,13]. Research in allergic rhinitis (AR) models shows that NLRP3 deficiency alleviates symptoms by enhancing mitophagy and reducing pyroptosis, highlighting its role in amplifying allergic responses [14,15]. Mitochondrial ROS-mediated NLRP3 activation has also been linked to allergic airway inflammation, underscoring a mechanistic pathway in hypersensitivity reactions [16,17]. Overall, NLRP3 appears to act as a sensor for allergens, contributing to chronic allergic diseases through sustained inflammation.
NLRP3 and Asthma:
In asthma, particularly neutrophilic and severe forms, NLRP3 drives airway hyperresponsiveness (AHR), eosinophilic infiltration, and cytokine production [18,19,20]. Studies in house dust mite (HDM)-induced models reveal that NLRP3 promotes pathogenesis in an inflammasome-dependent manner, with elevated expression correlating to steroid-resistant inflammation [21,22]. Mitochondrial ROS and endoplasmic reticulum stress further activate NLRP3, exacerbating chronic airway remodeling [16,23]. These data position NLRP3 as a central mediator in asthma’s inflammatory cascade, particularly in obesity-associated or allergen-driven subtypes.
NLRP3 and ADHD:
Evidence linking NLRP3 to ADHD is emerging but limited, primarily through associations with neuroinflammation and oxidative stress. Preclinical studies suggest that NLRP3 activation contributes to ADHD-like behaviors via microglial activation and TLR4 signaling [24,25,26]. Inhibition of NLRP3 has shown potential in ameliorating symptoms in models involving metabolic perturbations or excitotoxicity [27,28]. While direct causality remains understudied, NLRP3’s role in CNS inflammation may intersect with ADHD’s dopaminergic and glial dysregulation.
NLRP3 and Epilepsy:
NLRP3 is strongly implicated in epileptogenesis, where it promotes neuroinflammation, pyroptosis, and seizure progression through IL-1β release [29,30,31,32]. Mitochondrial dysfunction and ROS trigger NLRP3 in temporal lobe epilepsy, inhibiting mitophagy and exacerbating neuronal damage [33,34]. Studies in febrile infection-related epilepsy syndrome (FIRES) highlight microglial NLRP3 as a driver of protracted inflammation [35]. These findings suggest NLRP3 as a therapeutic target to mitigate seizure severity and cognitive deficits.
NLRP3 and Obesity:
NLRP3 activation in obesity fosters chronic low-grade inflammation, insulin resistance, and metabolic complications via sensing of danger signals like free fatty acids [36,37,38]. In adipose tissue, NLRP3 drives IL-1β production, correlating with obesity severity and atrial arrhythmias [39,40]. Enteric glial NLRP3 also impairs intestinal barrier function in obese models [41]. This positions NLRP3 at the nexus of obesity-induced systemic inflammation.
NLRP3 and Tourette’s Syndrome:
Links between NLRP3 and Tourette’s syndrome (TS) are sparse but suggest involvement in neurodevelopmental inflammation. Studies indicate NLRP3’s role in TS-like behaviors through pyroptosis and mitophagy dysregulation [24,42]. As part of broader neurodevelopmental disorders, NLRP3 may contribute via immune activation, though more research is needed to establish direct causality.
Two-Step Activation of NLRP3 and Threshold Dose Response to Aluminum:
NLRP3 inflammasome activation follows a canonical two-step process: priming (signal 1), which upregulates NLRP3 and pro-IL-1β via NF-κB signaling, and activation (signal 2), triggered by stressors like ATP or crystals, leading to assembly and cytokine maturation [43,44,45,46,47]. This biphasic mechanism implies a threshold effect, where inflammatory cytokines are only produced on the second exposure. Aluminum, used as a vaccine adjuvant, activates NLRP3 via lysosomal destabilization and ROS, mediated by receptors like GPRC6A [48]. The two step requirement creates a nonlinear dose response: the first dose primes without activating, while the second dose activates the inflammasome, triggering robust inflammation. This could explain variable responses to aluminum exposure in susceptible individuals, potentially linking to NLRP3-associated disorders.
Treatments and Interactions with NLRP3:
Therapeutic strategies targeting NLRP3 show promise across these conditions, often by inhibiting inflammasome assembly or downstream cytokines.
For ASD, salidroside ameliorates neuroinflammation by suppressing NLRP3-mediated pyroptosis, while phosphodiesterase 4 inhibitors reduce glial damage via NLRP3 modulation [49,50]. Acupuncture at ST36 also inhibits NLRP3, alleviating behavioral deficits [7].
In allergies, NLRP3 inhibitors like MCC950 or OLT1177 reduce inflammation and pyroptosis; natural compounds such as narirutin downregulate NLRP3 via USP15 inhibition [51,52,53]. IL-1β blockade offers complementary relief in allergic airway disease.
Asthma treatments include NLRP3 inhibitors like RRx-001, JT002, and MCC950, which attenuate AHR and steroid resistance [20,21,54]. Yupingfeng San and vitamin D3 suppress NLRP3, enhancing steroid sensitivity [55,56].
For ADHD, morin hydrate provides neuroprotection by targeting NLRP3 and TLR4; MCC950 shows potential in related neuroinflammatory models [25,27]. Bushen Kaiqiao Formula inhibits microglial NLRP3 activation [26].
Epilepsy interventions target NLRP3 with inhibitors like MCC950, reducing seizures and mitophagy impairment [29,31,57,58,59]. Klotho and cannabidiol alleviate inflammation via Nrf2 or CB2R pathways intersecting NLRP3 [60,61].
In obesity, colchicine and adiponectin receptor agonists suppress NLRP3-driven inflammation, improving insulin sensitivity [62,63]. Gut microbiota modulation and natural polyphenols like garlic chive vesicles inhibit NLRP3, reversing metabolic dysfunction [64,65,66].
For Tourette’s syndrome, Changpu Yujin Tang mitigates symptoms by inhibiting NLRP3 via enhanced mitophagy, representing a novel herbal approach [43].
In summary, NLRP3 emerges as a convergent pathway in these disorders along with potentially explaining nonlinearities seen at the population level.

5. Conclusions

This study leverages population-level data from the NSCH 2020-2023 and CDC NIS 2011-2017 to reveal significant associations between aluminum-adjuvanted vaccine uptake during specific early childhood periods and the prevalence of inflammatory conditions such as autism, allergies, ADHD, asthma, epilepsy, obesity, and Tourette’s syndrome across U.S. states. Notably, these associations exhibit nonlinear patterns, including threshold-like responses and sign reversals across age windows, which align with the two-step priming and activation mechanism of the NLRP3 inflammasome, a pathway implicated in each of these disorders through neuroinflammation, oxidative stress, and immune dysregulation. In contrast, blood disorders, serving as a negative control, showed no such relationships, underscoring the specificity of these findings to inflammatory outcomes.
The evidence presented supports the hypothesis that aluminum adjuvants, present in vaccines like DTaP, HepB, Hib, PCV, and Polio, may contribute to these conditions via NLRP3-mediated inflammation, particularly in susceptible populations. Inter-disease correlations further suggest shared inflammatory underpinnings, while vaccine-specific breakdowns challenge alternative explanations like hidden confounders. These results extend prior mechanistic studies on NLRP3’s role in disease pathogenesis and highlight promising therapeutic avenues, including NLRP3 inhibitors (e.g., MCC950), natural compounds, and lifestyle interventions that could mitigate risks.
While ecological in nature, these findings warrant urgent prospective studies to confirm causality, refine vaccination schedules for minimized aluminum exposure, and explore genetic or environmental modifiers of NLRP3 sensitivity. Ultimately, prioritizing inflammasome-targeted research could inform safer immunization strategies and novel treatments, reducing the burden of these chronic childhood conditions on individuals and society.

Acknowledgments

This study was self funded, there are no conflicts of interest, and I am the sole author.

Appendix

Code and data used:

References

  1. Child and Adolescent Health Measurement Initiative (CAHMI) (2020-2021). 2020-2021 National Survey of Children's Health, CSV Indicator dataset. Data Resource Center for Child and Adolescent Health supported by Cooperative Agreement U59MC27866 from the U.S. Department of Health and Human Services, Health Resources and Services Administration (HRSA), Maternal and Child Health Bureau (MCHB). Retrieved 02/01/23 from childhealthdata.
  2. Child and Adolescent Health Measurement Initiative (CAHMI) (2025). 2022-2023 NationalSurvey of Children’s Health. SPSS codebook for data users: Child and Family HealthMeasures, National Performance and Outcome Measures, and Subgroups, Version 1.0. DataResource Center for Child and Adolescent Health supported by Cooperative AgreementU59MC27866 from the U.S. Department of Health and Human Services, Health Resourcesand Services Administration (HRSA), Maternal and Child Health Bureau (MCHB). Retrieved[02/01/25] from childhealthdata.
  3. U.S. Department of Health and Human Services (DHHS). National Center for Immunization and Respiratory Diseases. The 2011-2017 National Immunization Survey - Child. Atlanta, GA: Centers for Disease Control and Prevention, 2022.
  4. Mokeddem, K. (2025). Age at Vaccination a Potential Confounder of Studies of Aluminum Adjuvants. Preprints. [CrossRef]
  5. Vallese, A. , et al. NLRP3 inflammasome-mitochondrion loop in autism spectrum disorder. Free Radical Biology and Medicine 2024, 225, 581–594. [Google Scholar] [CrossRef]
  6. Saresella, M. , et al. Multiple inflammasome complexes are activated in autistic spectrum disorders. Brain, Behavior, and Immunity 2016, 57, 125–133. [Google Scholar] [CrossRef] [PubMed]
  7. Zhao, P. , Li, X., Wang, Y., Zhang, H., & Liu, H. Acupuncture at ST36 Alleviates the Behavioral Disorder of Autistic Rats by Inhibiting the TXNIP-NLRP3 Inflammasome Pathway. Journal of Neuropathology & Experimental Neurology 2022, 81, 127–134. [Google Scholar] [CrossRef]
  8. Szabo, A. , et al. Elevated levels of peripheral and central nervous system immune markers reflect innate immune dysregulation in autism spectrum disorder. Psychiatry Research 2024, 342, 116245. [Google Scholar] [CrossRef]
  9. Jung, H. , et al. The NLRP3 inflammasome in microglia regulates repetitive behavior by modulating NMDA glutamate receptor functions. Cell Reports 2025, 44, 115656. [Google Scholar] [CrossRef] [PubMed]
  10. Hylén, U. , et al. Potential transdiagnostic lipid mediators of inflammatory activity in individuals with serious mental illness. Frontiers in Psychiatry 2021, 12, 778325. [Google Scholar] [CrossRef]
  11. Lu, H.-F. , Zhou, Y.-C., Hu, T.-Y., Yang, D.-H., Wang, X.-J., Luo, D.-D., Qiu, S.-Q., & Cheng, B.-H. Unraveling the role of NLRP3 inflammasome in allergic inflammation: implications for novel therapies. Frontiers in Immunology 2024, 15, 1435892. [Google Scholar] [CrossRef]
  12. Zhou, H. , Wang, L., Lv, W., & Yu, H. The NLRP3 inflammasome in allergic diseases: mechanisms and therapeutic implications. Clinical and Experimental Medicine 2024, 24, 231. [Google Scholar] [CrossRef]
  13. Xiao, Y. , Li, X., & Li, D. NLRP3 inflammasome: A likely target for the treatment of allergic diseases. Clinical and Experimental Allergy 2018, 48, 1080–1091. [Google Scholar] [CrossRef]
  14. Ding, H. , et al. NLRP3 inflammasome deficiency alleviates inflammation and oxidative stress by promoting PINK1/Parkin-mediated mitophagy in allergic rhinitis mice and nasal epithelial cells. Journal of Asthma and Allergy 2024, 17, 717–731. [Google Scholar] [CrossRef]
  15. Yue, J. , et al. NLRP3 inflammasome activation promotes the development of allergic rhinitis via epithelium pyroptosis. Biochemical and Biophysical Research Communications 2020, 521, 1051–1058. [Google Scholar] [CrossRef]
  16. Kim, S. R. , Kim, D. I., Kim, S. H., Lee, H., Lee, K. S., Cho, S. H., & Lee, Y. C. NLRP3 inflammasome activation by mitochondrial ROS in bronchial epithelial cells is required for allergic inflammation. Cell Death & Disease 2014, 5, e1498. [Google Scholar] [CrossRef]
  17. Leszczyńska, K. , et al. The NLRP3 inflammasome as a new target in respiratory disorders treatment. Frontiers in Immunology 2022, 13, 1006654. [Google Scholar] [CrossRef] [PubMed]
  18. Williams, E. J. , et al. Role of the NLRP3 inflammasome in asthma: Relationship with neutrophilic inflammation, obesity, and therapeutic options. Journal of Allergy and Clinical Immunology 2021, 147, 2060–2062. [Google Scholar] [CrossRef] [PubMed]
  19. Theofani, E. , et al. Targeting NLRP3 inflammasome activation in severe asthma. Journal of Clinical Medicine 2019, 8, 1615. [Google Scholar] [CrossRef] [PubMed]
  20. Kim, R. Y. , et al. Role for NLRP3 inflammasome-mediated, IL-1β-dependent responses in severe, steroid-resistant asthma. American Journal of Respiratory and Critical Care Medicine 2017, 196, 283–297. [Google Scholar] [CrossRef]
  21. Ma, M. , Li, G., Qi, M., Jiang, W., & Zhou, R. Inhibition of the inflammasome activity of NLRP3 attenuates HDM-induced allergic asthma. Frontiers in Immunology 2021, 12, 718779. [Google Scholar] [CrossRef]
  22. Simpson, J. L. , & Powell, N. Elevated expression of the NLRP3 inflammasome in neutrophilic asthma. European Respiratory Journal 2014, 43, 1067–1076. [Google Scholar] [CrossRef]
  23. [Wu, Y. , Di, X., Zhao, M., Li, H., Bai, L., & Wang, K. The role of the NLRP3 inflammasome in chronic inflammation in asthma and chronic obstructive pulmonary disease. Immun Inflamm Dis 2022, 10, e750. [Google Scholar] [CrossRef]
  24. Shahi, A. , et al. The relationship between neurodevelopmental disorders (NDDs) and NLRP3 inflammasome. Inflammation Research 2025, 74, 90. [Google Scholar] [CrossRef]
  25. Salem, H. A. , Abdelaziz, S. A. M., Eldaim, M. A. A., AbdelDaim, M. M., & Elgarawany, G. E. Neuroprotective effect of morin hydrate against attention-deficit/hyperactivity disorder (ADHD) induced by MSG and/or protein malnutrition in rat pups: Effect on oxidative/monoamines/inflammatory balance and apoptosis. Pharmaceuticals 2022, 15, 1012. [Google Scholar] [CrossRef]
  26. Zhang, J. , et al. A study on the mechanism of Bushen Kaiqiao Formula in modulating microglial activation to alleviate neuroinflammation in ADHD. Biochem Biophys Rep 2025, 43, 102211. [Google Scholar] [CrossRef]
  27. Chen, K.-P. , Wang, Y.-C., Lin, C.-H., Chen, C.-C., Lin, C.-C., & Chen, S.-Y. A selective inhibitor of the NLRP3 inflammasome as a potential therapeutic approach for neuroprotection in a transgenic mouse model of Huntington's disease. Journal of Neuroinflammation 2022, 19, 56. [Google Scholar] [CrossRef] [PubMed]
  28. Kovács, Z. , Thavendiranathan, P., & Wicks, S. Therapeutic potential of exogenous ketone supplement induced ketosis in the treatment of psychiatric disorders: Review of current literature. Frontiers in Psychiatry 2019, 10, 363. [Google Scholar] [CrossRef]
  29. Chen, J. , et al. Mechanism of NLRP3 inflammasome in epilepsy and related therapeutic agents. Neuroscience 2024, 546, 157–177. [Google Scholar] [CrossRef]
  30. Palumbo, L. , et al. The NLRP3 inflammasome in neurodegenerative disorders: Insights from epileptic models. Biomedicines 2023, 11, 2825. [Google Scholar] [CrossRef]
  31. Yue, J. , Wei, Y. J., Yang, X. L., Liu, S. Y., Yang, H., & Zhang, C.-Q. NLRP3 inflammasome and endoplasmic reticulum stress in the epileptogenic zone in temporal lobe epilepsy: Molecular insights into their interdependence. Neuropathology and Applied Neurobiology 2020, 46, 770–785. [Google Scholar] [CrossRef] [PubMed]
  32. Mohseni-Moghaddam, P. , et al. A literature overview on epilepsy and inflammasome activation. Brain Research Bulletin 2021, 172, 229–235. [Google Scholar] [CrossRef] [PubMed]
  33. Khedpande, N. , et al. (2025). Mitochondrial dysfunction and NLRP3 inflammasome activation in drug-resistant epilepsy: emerging insights and mitochondrial-targeted therapeutic strategies. Naunyn-Schmiedeberg's Archives of Pharmacology. Advance online publication. [CrossRef]
  34. Wu, M. , et al. NLRP3 inflammasome inhibits mitophagy during the progression of temporal lobe epilepsy. Sci Rep 2025, 15, 16341. [Google Scholar] [CrossRef]
  35. Lin, W.-S. , et al. Hypothesis: Febrile infection-related epilepsy syndrome is a microglial NLRP3 inflammasome/IL-1 axis-driven autoinflammatory syndrome. Clinical & Translational Immunology 2021, 10, e1299. [Google Scholar] [CrossRef]
  36. Vandanmagsar, B. , Youle, R. J., & Sag, D. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nature Medicine 2011, 17, 179–188. [Google Scholar] [CrossRef]
  37. Jorquera, G. , López-Alarcón, C., & Brandan, E. NLRP3 inflammasome: Potential role in obesity related low-grade inflammation and insulin resistance in skeletal muscle. International Journal of Molecular Sciences 2021, 22, 3254. [Google Scholar] [CrossRef]
  38. Kaiser, W. , Ahmad, S., Kumar, A., & Ahmad, F. Role of NLRP3 inflammasome activation in obesity-mediated metabolic disorders. International Journal of Environmental Research and Public Health 2021, 18, 511. [Google Scholar] [CrossRef]
  39. Scott, L., Jr. , et al. NLRP3 inflammasome is a key driver of obesity-induced atrial arrhythmias. Cardiovascular Research 2021, 117, 1746–1759. [Google Scholar] [CrossRef]
  40. Rheinheimer, J. , et al. Current role of the NLRP3 inflammasome on obesity and insulin resistance: A systematic review. Metabolism 2017, 74, 1–9. [Google Scholar] [CrossRef]
  41. D'Antongiovanni, V. , Pellegrini, C., Fornai, M., et al. Enteric glial NLRP3 inflammasome contributes to gut mucosal barrier alterations in a mouse model of diet-induced obesity. Acta Physiologica 2025, 241, e14232. [Google Scholar] [CrossRef]
  42. Huang, S. , et al. Changpu Yujin Tang mitigates tourette syndrome by enhancing PINK1/Parkin-mediated mitophagy and suppressing NLRP3 inflammasome-mediated pyroptosis. Immunobiology 2025, 230, 153118. [Google Scholar] [CrossRef]
  43. Swanson, K. V. , Deng, M., & Ting, J. P.-Y. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nature Reviews Immunology 2019, 19, 477–489. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, Y. , Wang, H., Kouadir, M. et al. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis 2019, 10, 128. [Google Scholar] [CrossRef] [PubMed]
  45. Blevins, H. M. , Xu, Y., Biby, S., & Zhang, S. The NLRP3 inflammasome pathway: A review of mechanisms and inhibitors for the treatment of inflammatory diseases. Frontiers in Aging Neuroscience 2022, 14, 879021. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, Y. , Ye, X., Escames, G. et al. The NLRP3 inflammasome: contributions to inflammation-related diseases. Cell Mol Biol Lett 2023, 28, 51. [Google Scholar] [CrossRef]
  47. Huang, W. , Zhang, Z., Qiu, Y., Gao, Y., Fan, Y., Wang, Q., & Zhou, Q. NLRP3 inflammasome activation in response to metals. Frontiers in Immunology 2023, 14, 1055788. [Google Scholar] [CrossRef]
  48. Quandt, D. , Wiedemann, N., Frick, M., Kabelitz, D., & Alkhaled, S. GPRC6A mediates Alum-induced Nlrp3 inflammasome activation but limits Th2 type antibody responses. Scientific Reports 2015, 5, 16719. [Google Scholar] [CrossRef]
  49. Wu, Q. , et al. Salidroside ameliorates neuroinflammation in autistic rats by inhibiting NLRP3/Caspase-1/GSDMD signal pathway. Brain Res Bull 2025, 220, 111132. [Google Scholar] [CrossRef]
  50. Adili, A. , et al. Research progress on phosphodiesterase 4 inhibitors in central nervous system diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024, 53, 390–398. [Google Scholar] [CrossRef] [PubMed]
  51. Shih, M.-C. , Chen, C.-Y., & Lin, C.-C. Inhibition of NLRP3 inflammasome activation by 3H-1,2-dithiole-3-thione: A potential therapeutic approach for psoriasis treatment. International Journal of Molecular Sciences 2023, 24, 13528. [Google Scholar] [CrossRef]
  52. Yang, J. , et al. Narirutin downregulates USP15 to inhibit the NLRP3 ubiquitination in HDM-induced allergic rhinitis. Phytomedicine 2025, 144, 156895. [Google Scholar] [CrossRef]
  53. Besnard, A. G. , et al. Functional relevance of NLRP3 inflammasome-mediated interleukin (IL)-1β during acute allergic airway inflammation. Clinical and Experimental Immunology 2014, 178, 212–223. [Google Scholar] [CrossRef]
  54. Ambrus-Aikelin, G. , et al. JT002, a small molecule inhibitor of the NLRP3 inflammasome for the treatment of autoinflammatory disorders. Sci Rep 2023, 13, 13524. [Google Scholar] [CrossRef]
  55. Liu, X. , Shen, J., Fan, D., Qiu, X., Guo, Q., Zheng, K., Luo, H., Shu, J., Lu, C., Zhang, G., Lu, A., Ma, C., & He, X. Yupingfeng San inhibits NLRP3 inflammasome to attenuate the inflammatory response in asthma mice. Frontiers in Pharmacology 2017, 8, 944. [Google Scholar] [CrossRef]
  56. Khalil, B. , et al. Vitamin D3 suppresses NLRP3 inflammasome pathway and enhances steroid sensitivity in a neutrophilic steroid hyporesponsive asthma mouse model. Inflammation Research 2025, 74, 51. [Google Scholar] [CrossRef]
  57. Haque, I. , Thapa, P., Burns, D. M., Zhou, J., Sharma, M., Sharma, R., & Singh, V. NLRP3 inflammasome inhibitors for antiepileptogenic drug discovery and development. International Journal of Molecular Sciences 2024, 25, 6078. [Google Scholar] [CrossRef]
  58. Zulfat, M. , et al. Identification of novel NLRP3 inhibitors as therapeutic options for epilepsy by machine learning-based virtual screening, molecular docking and biomolecular simulation studies. Heliyon 2024, 10, e34410. [Google Scholar] [CrossRef]
  59. Foiadelli, T. , Santangelo, A., Costagliola, G., Costa, E., Scacciati, M., Riva, A., Volpedo, G., Smaldone, M., Bonuccelli, A., Clemente, A. M., Ferretti, A., Savasta, S., Striano, P., & Orsini, A. Neuroinflammation and status epilepticus: a narrative review unraveling a complex interplay. Frontiers in Pediatrics 2023, 11, 1251914. [Google Scholar] [CrossRef]
  60. Xiang, T. , et al. Klotho alleviates NLRP3 inflammasome-mediated neuroinflammation in a temporal lobe epilepsy rat model by activating the Nrf2 signaling pathway. Epilepsy Behav 2022, 128, 108509. [Google Scholar] [CrossRef]
  61. Cai, Y. , et al. Cannabinoid receptor 2 agonist AM1241 alleviates epileptic seizures and epilepsy-associated depression via inhibiting neuroinflammation in a pilocarpine-induced chronic epilepsy mouse model. Molecular and Cellular Neuroscience 2024, 130, 103958. [Google Scholar] [CrossRef]
  62. Demidowich, A. P. , et al. Colchicine to decrease NLRP3-activated inflammation and improve obesity-related metabolic dysregulation. Medical Hypotheses 2016, 92, 67–73. [Google Scholar] [CrossRef] [PubMed]
  63. Li G, Huang D, Kidd JM, Zou Y, Wu X, Zhang Y, Gehr TWB, Li N, Li PL. Acid ceramidase as a novel target for adiponectin receptor agonist to abrogate podocyte NLRP3 inflammasome activation and glomerular inflammation during obesity. J Pharmacol Exp Ther. 2025, 392, 103757. [Google Scholar] [CrossRef] [PubMed]
  64. Huang, H. , Zhou, T., He, F., Wen, B., Yang, Y., Zhong, W., Wang, Q., & Li, J. The gut microbiota improves reproductive dysfunction in obese mice by suppressing the NLRP3/ASC/caspase-1 axis. Future Microbiology 2024, 19, 1389–1405. [Google Scholar] [CrossRef] [PubMed]
  65. Liu, B. , et al. Therapeutic potential of garlic chive-derived vesicle-like nanoparticles in NLRP3 inflammasome-mediated inflammatory diseases. Theranostics 2021, 11, 9311–9330. [Google Scholar] [CrossRef] [PubMed]
  66. Wang T, Xu H, Dong R, Wu S, Guo Y, Wang D. Effectiveness of targeting the NLRP3 inflammasome by using natural polyphenols: A systematic review of implications on health effects. Food Res Int. 2023, 165, 112567. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated