Neuroimmune mechanisms of cognitive impairment in a mouse model of Gulf War illness

Author(s):  
Joshua D. Bryant ◽  
Maheedhar Kodali ◽  
Bing Shuai ◽  
Saeed S. Menissy ◽  
Paige J. Graves ◽  
...  
2020 ◽  
Author(s):  
Joshua D. Bryant ◽  
Maheedhar Kodali ◽  
Bing Shuai ◽  
Saeed S. Menissy ◽  
Paige J. Graves ◽  
...  

AbstractGulf War Illness (GWI) is a chronic, multi-symptom disorder affecting approximately 30 percent of the nearly 700,000 veterans of the 1991 Persian Gulf War. Recent studies have revealed that GWI-related chemical (GWIC) exposure promotes immune activation and metabolic rewiring, which correlate with neurocognitive impairments and other symptoms of GWI. However, the molecular mechanisms and signaling pathways linking GWIC to inflammation, metabolic alterations, and neurological symptoms remain unclear. Mitochondrial dysfunction has been documented in veterans with GWI and rodent models, and because mitochondria are key immune regulators, we hypothesized that alterations to mitochondria-immune crosstalk could contribute to the development of GWI-related symptoms. Here we show that acute exposure of murine macrophages to GWIC alters mitochondrial respiration and potentiates innate immune signaling and inflammatory cytokine secretion. Using an established mouse model of GWI, we report that neurobehavioral changes, neuroinflammation, and mitochondrial protein rewiring are attenuated in mice lacking the cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) and NOD-, LRR- or pyrin domain-containing protein 3 (NLRP3) innate immune pathways. Finally, we report sex differences in response to GWIC, with female mice showing more pronounced cognitive impairment, neuroinflammation, and mitochondrial protein alterations in the brain compared to male mice. Our results provide novel information on sex differences in this model and suggest that STING and NLRP3 are key mediators of the cognitive impairment, inflammation, and mitochondrial dysfunction observed in GWI.


Life Sciences ◽  
2021 ◽  
pp. 119675
Author(s):  
Mariana Angoa-Pérez ◽  
Branislava Zagorac ◽  
Dina M. Francescutti ◽  
Kevin R. Theis ◽  
Donald M. Kuhn

Life Sciences ◽  
2021 ◽  
pp. 119707
Author(s):  
Kyle A. Brown ◽  
Jessica M. Carpenter ◽  
Collin J. Preston ◽  
Helaina D. Ludwig ◽  
Kendall B. Clay ◽  
...  

2019 ◽  
Vol 70 ◽  
pp. 26-32 ◽  
Author(s):  
Lindsay T. Michalovicz ◽  
Alicia R. Locker ◽  
Kimberly A. Kelly ◽  
Julie V. Miller ◽  
Zachary Barnes ◽  
...  

2020 ◽  
Vol 14 ◽  
Author(s):  
Jun Gao ◽  
Fuyi Xu ◽  
Athena Starlard-Davenport ◽  
Diane B. Miller ◽  
James P. O’Callaghan ◽  
...  

PLoS ONE ◽  
2015 ◽  
Vol 10 (3) ◽  
pp. e0119579 ◽  
Author(s):  
Zuchra Zakirova ◽  
Miles Tweed ◽  
Gogce Crynen ◽  
Jon Reed ◽  
Laila Abdullah ◽  
...  

Author(s):  
Zuchra Zakirova ◽  
Gogce Crynen ◽  
Samira Hassan ◽  
Laila Abdullah ◽  
Lauren Horne ◽  
...  

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Utsav Joshi ◽  
James E. Evans ◽  
Ross Joseph ◽  
Tanja Emmerich ◽  
Nicole Saltiel ◽  
...  

2021 ◽  
Vol 11 (7) ◽  
pp. 905
Author(s):  
Punnag Saha ◽  
Peter T. Skidmore ◽  
LaRinda A. Holland ◽  
Ayan Mondal ◽  
Dipro Bose ◽  
...  

Gulf War Illness (GWI) is a chronic multi-symptomatic illness that is associated with fatigue, pain, cognitive deficits, and gastrointestinal disturbances and presents a significant challenge to treat in clinics. Our previous studies show a role of an altered Gut–Brain axis pathology in disease development and symptom persistence in GWI. The present study utilizes a mouse model of GWI to study the role of a labdane diterpenoid andrographolide (AG) to attenuate the Gut–Brain axis-linked pathology. Results showed that AG treatment in mice (100 mg/kg) via oral gavage restored bacteriome alterations, significantly increased probiotic bacteria Akkermansia, Lachnospiraceae, and Bifidobacterium, the genera that are known to aid in preserving gut and immune health. AG also corrected an altered virome with significant decreases in virome families Siphoviridae and Myoviridae known to be associated with gastrointestinal pathology. AG treatment significantly restored tight junction proteins that correlated well with decreased intestinal proinflammatory mediators IL-1β and IL-6 release. AG treatment could restore Claudin-5 levels, crucial for maintaining the BBB integrity. Notably, AG could decrease microglial activation and increase neurotrophic factor BDNF, the key to neurogenesis. Mechanistically, microglial conditioned medium generated from IL-6 stimulation with or without AG in a concentration similar to circulating levels found in the GWI mouse model and co-incubated with neuronal cells in vitro, decreased Tau phosphorylation and neuronal apoptosis. In conclusion, we show that AG treatment mitigated the Gut–Brain-Axis associated pathology in GWI and may be considered as a potential therapeutic avenue for the much-needed bench to bedside strategies in GWI.


Sign in / Sign up

Export Citation Format

Share Document