Microglial Immunometabolism Endophenotypes Implicated in Sex Differences in Alzheimer’s Disease

Study suggests sex-specific pathways show potential for sex-specific therapeutic approaches

23-NEU-4390509-CQD-Hero-650×450

Sex-related differences in Alzheimer’s disease (AD) pathogenesis and disease progression may be due in part to sex-specific differences in immune responses, cellular metabolism and microglial immunometabolism, according to new Cleveland Clinic research. The findings, published in Alzheimer’s and Dementia, offer important insights for the development of sex-specific treatment and prevention options for AD.

Advertisement

Cleveland Clinic is a non-profit academic medical center. Advertising on our site helps support our mission. We do not endorse non-Cleveland Clinic products or services. Policy

“Women are more likely to develop Alzheimer’s disease, and they experience faster cognitive decline compared with their male counterparts,” says Feixiong Cheng, PhD, corresponding author of the research report and associate staff in Cleveland Clinic’s Genomic Medicine Institute. “Yet how cell type-specific immune responses and cellular mechanisms mediate the apparent sex differences in Alzheimer’s pathophysiology has been unclear.”

“The immune system depends on communication between different cell types fueled by energy created from unique metabolic processes,” adds co-author Justin Lathia, PhD, Vice Chair of the Department of Cardiovascular and Metabolic Sciences in Cleveland Clinic’s Lerner Research Institute. “As sex influences both the immune system and metabolic processes, our study aimed to identify how all of these individual factors influence one another to contribute to Alzheimer’s disease.”

“We hypothesized that an interplay exists between cellular metabolism and immune systems,” Dr. Cheng explains, “and that understanding this interplay, which is termed the ‘immunometabolism endophenotype,’ will be essential for improving understanding of sex differences in Alzheimer’s pathogenesis and developing individualized treatments in a sex-specific manner.”

The study in brief

The study was funded in part by a grant from the National Institutes of Health supporting the use of systems biology and multi-omics approaches to untangle the complex interactions between the immune system, inflammation, and sex-based differences in AD. It is part of an ongoing effort between investigators with Lerner Research Institute and the Cleveland Clinic Lou Ruvo Center for Brain Health to further understand the link between sex and various neurodegenerative conditions.

Advertisement

The researchers analyzed publicly available sequencing data obtained from the brains of 469 individuals, including 203 with AD, 117 with mild cognitive impairment, 7 with other forms of dementia and 142 controls with no cognitive impairment. They looked for changes in gene expression that indicated differences in immune function, cellular metabolism and signaling patterns between different cells in the brain. Results indicated sex-specific differences in all three of these factors in the brains of male versus female AD patients, especially in microglial cells ― the immune cells that reside in the brain.

Specific findings included the following:

  • Sex-specific immune metabolites, gene networks and signaling pathways were associated with AD pathogenesis and progression.
  • Females with AD exhibited microglial immunometabolism endophenotypes marked by reduced glutamate metabolism and enhanced interleukin-10 pathway activity.
  • Females with AD demonstrated a shift in glutamate-mediated cell-cell communications between excitatory neurons and microglia and astrocytes.

“Our results point to new understanding of the molecular basis for female predominance in AD,” the researchers conclude in their study report. They add that their findings call for independent validation in ethnically diverse cohorts to assess “a likely causal relationship of microglial immunometabolism in the sex differences in AD.”

Findings in context

“The more progress we make into measures to protect against Alzheimer’s disease, the more we see that it can’t be a one-size-fits-all approach,” Dr. Cheng says. “This project was about figuring out how these various individual established contributors to Alzheimer’s disease fit together. Once we determine that, we can design targeted treatments to prevent the disease or halt its progression.”

Advertisement

The project is one of multiple research avenues Dr. Cheng’s lab is pursuing with multidisciplinary collaborators at Cleveland Clinic and beyond to understand and overcome AD on various fronts. This collective work, which has drawn more than $16 million in external grant support in 2023, is profiled in a recent Consult QD article.

“At its core, Alzheimer’s disease is a woman’s health issue,” notes one of Dr. Cheng’s collaborators, Jessica Caldwell, PhD, Director of the Women’s Alzheimer’s Movement Prevention Center at Cleveland Clinic and a co-author on the current study. “To address it, we must understand how biological sex contributes to the underpinnings of this disease. These findings confirm that we need to look at sex differences in the way the body and brain systems communicate to be able to truly offer women personalized care. We look forward to continuing this research.”

Yuan Hou, PhD, a research associate with the Cheng lab, is the first author of this study, which was supported by the National Institute on Aging (R01AG084250) under the National Institutes of Health.

Related Articles

gut microbes in intestine
Cleveland Clinic, Tufts University Research Ties Gut Microbial TMAO Pathway to Chronic Kidney Disease

Large-scale joint study links elevated TMAO blood levels and chronic kidney disease risk over time

patient in ICU
Cleveland Clinic and Purdue Seek to Revolutionize Intensive Care Through AI

Investigators are developing a deep learning model to predict health outcomes in ICUs.

24-NEU-4528160-genetics-parkinson-disease-650×450
Multi-Ancestry Genetic Study of Parkinson’s Disease Identifies New Risk Genes in Pursuit of Novel Treatment Targets

International collaboration is most genetically diverse study of the disease to date

23-NEU-4357266-stock-brain-image_650x450
Noninvasive Technology Enhances Ability to Map Brain Activity to Track Behavior Change

Preclinical work promises large-scale data with minimal bias to inform development of clinical tests

23-NEU-4189360-hydrogen-sulfide-650×450
Can Boosting Hydrogen Sulfide Bolster Standard-of-Care Glioblastoma Therapy to Extend Survival?

Cleveland Clinic researchers pursue answers on basic science and clinical fronts

23-CCC-4375928 Quantum Innovation Catalyzer 650×450
A Unique Opportunity to Explore Quantum Computing’s Potential

Cleveland Clinic launches Quantum Innovation Catalyzer Program to help start-up companies access advanced research technology

Light trails coming from African American’s head
Blood-Based Biomarkers for Alzheimer’s Disease in Women (Podcast)

Research project aims to pinpoint biomarkers that could speed diagnosis

23-NEU-4216627_researcher-in-lab_650x450
An All-Fronts Approach to Understanding and Overcoming Alzheimer’s Disease

A conversation with Feixiong Cheng, PhD, about his wide-ranging research initiatives

Ad