Selected Grantee Publications
- Clear All
- 92 results found
- COVID-19/Coronavirus
- Microbiome
- Women's Health
Effect of Omeprazole on Esophageal Microbiota in Dogs Detected Using a Minimally Invasive Sampling Method
Handa et al., Journal of Veterinary Internal Medicine. 2025.
https://pmc.ncbi.nlm.nih.gov/articles/PMC11864821
Omeprazole alters the esophageal microbiome (EM) of humans and has associated effects. The changes and subsequent recovery of the EM in 3- to 6-year-old dogs after omeprazole treatment were assessed using the esophageal string test (EST). All 10 dogs tolerated the EST without adverse effects, and the EST retrieved sufficient biofluid to characterize the EM. Diversity analysis revealed no significant alterations in alpha (Observed species, Shannon and Simpson indices) and beta diversity (Bray‐Curtis) across the time points after omeprazole administration. Thus, omeprazole therapy was not observed to alter the EM of healthy dogs in this study. The application of EST in dogs illustrates its use as a minimally invasive tool for investigating the role of EM in esophageal health and disease in dogs. Supported by ORIP (K01OD030515).
Senescent-like Microglia Limit Remyelination Through the Senescence Associated Secretory Phenotype
Gross et al., Nature Communications. 2025.
https://www.nature.com/articles/s41467-025-57632-w
Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disease in which immune cells infiltrate the central nervous system and promote deterioration of myelin and neurodegeneration. The capacity to regenerate myelin in the central nervous system diminishes with age. In this study, researchers used 2- to 3-month-old (young), 12-month-old (middle-aged), and 18- to 22-month-old (aged) C57BL/6 male and female mice. Results showed an upregulation of the senescence marker P16ink4a (P16) in microglial and macrophage cells within demyelinated lesions. Notably, treatment of senescent cells using genetic and pharmacological senolytic methods leads to enhanced remyelination in young and middle-aged mice but fails to improve remyelination in aged mice. These results suggest that therapeutic targeting of senescence-associated secretory phenotype components may improve remyelination in aging and MS. Supported by ORIP (R24OD036199), NIA, NINDS, and NIMH.
Local Tissue Response to a C-X-C Motif Chemokine Ligand 12 Therapy for Fecal Incontinence in a Rabbit Model
Ruetten et al., American Journal of Physiology—Gastrointestinal and Liver Physiology. 2025.
https://pubmed.ncbi.nlm.nih.gov/39745592
Obstetric anal sphincter injury (OASI) occurs in 2–7% of vaginal childbirths. Surgical interventions for OASI are suboptimal, with 30% of women reporting continued reduction in quality of life due to long-term fecal incontinence. Researchers used a 4- to 5-month-old female New Zealand white rabbit model for OASI to determine whether local C-X-C motif chemokine ligand 12 (CXCL12) injection reduces postinjury pathologies. Treatment with CXCL12 significantly reduced fibrosis. Untreated rabbits demonstrated reduced distinction of anal sphincter skeletal muscle layering and significantly increased the amount of fibrosis. Treatment with CXCL12 did not affect recruitment of CD34+ cells, the number of PAX7+ satellite cells, or innervation and vascularization of skeletal muscle. This pilot study demonstrates the potential of a novel therapeutic for OASI. Supported by ORIP (T32OD010957).
Failure of Colonization Following Gut Microbiota Transfer Exacerbates DSS-Induced Colitis
Gustafson et al., Gut Microbes. 2025.
https://pubmed.ncbi.nlm.nih.gov/39812347/
Microorganisms that inhabit the gastrointestinal tract, known as the gut microbiome (GM), play a vital role in health and disease. Dysbiosis, the reduced richness of symbiotic commensals in the GM, exacerbates inflammation and increases inflammatory bowel disease (IBD) severity. Researchers used a mouse model for IBD to determine the role of GM composition, richness, and transfer methods on IBD disease severity. A comparison of GM transfer methods demonstrated that co-housing was not as efficient as embryonic transfer and cross-fostering. The GM of the donor and recipient during co-housing determined transfer efficiency. Transfer of a low richness GM to a recipient with high GM richness, followed by dextran sodium sulfate administration to induce IBD, resulted in significant weight loss, greater lesion severity, increased inflammatory response, and higher mortality rates. This study provides evidence regarding the role of GM composition and colonization in IBD modulation. Supported by ORIP (T32OD011126, U42OD010918) and NIGMS.
Pre-Challenge Gut Microbial Signature Predicts RhCMV/SIV Vaccine Efficacy in Rhesus Macaques
Brochu et al., Microbiology Spectrum. 2025.
https://journals.asm.org/doi/10.1128/spectrum.01285-24
Rhesus cytomegalovirus–based simian immunodeficiency virus (RhCMV/SIV) vaccines provide protection against SIV challenge in approximately 60% of vaccinated rhesus macaques. This study assessed the role that gut microbiota play in SIV vaccine efficacy by analyzing the microbiomes of rhesus macaques before and after immunization using novel compositional data analysis techniques and machine-learning strategies. Researchers identified a gut microbial signature that predicted vaccine protection outcomes and correlated with early biomarker changes in the blood (i.e., host immune response to vaccination). This study indicates that the gut microbiome might play a role in vaccine-induced immunity. Supported by ORIP (P51OD011092).
Integrative Multi-omics Analysis Uncovers Tumor-Immune-Gut Axis Influencing Immunotherapy Outcomes in Ovarian Cancer
Rosario et al., Nature Communications. 2024.
https://pubmed.ncbi.nlm.nih.gov/39638782
Recurrent ovarian cancer (OC) is the deadliest gynecological malignancy, with a 5-year survival rate of 50% and a median progression-free survival (PFS) of 1.9 to 2.1 months. A trial cohort of 40 patients was treated with a combination of the anti-PD-1 pembrolizumab, the anti–vascular endothelial growth factor bevacizumab, and cyclophosphamide. The investigators conducted a multi-omics analysis—including transcriptomic analysis, digital spatial profiling, 16s-rRNA sequencing, and metabolomics—to understand the underlying mechanisms for the enhanced PFS to a median of 10.2 months and overall response rate of 47.5%. Multi-omics analysis highlighted the formation of tertiary lymphoid structures known to improve responses to immunotherapy, differential microbial patterns, and alterations in the metabolites in three key metabolism pathways that enhanced immune response in patients to produce a durable clinical response. These findings highlight the importance of the tumor microenvironment and the gut microbiome, along with its metabolites, in elevating the efficacy of the cocktail therapy in recurrent OC patients, thereby enhancing their survival and quality of life. Supported by ORIP (S10OD024973) and NCI.
A Single-Dose Intranasal Live-Attenuated Codon Deoptimized Vaccine Provides Broad Protection Against SARS-CoV-2 and Its Variants
Liu et al., Nature Communications. 2024.
https://pubmed.ncbi.nlm.nih.gov/39187479
Researchers developed an intranasal, single-dose, live-attenuated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) vaccine (CDO-7N-1) using codon deoptimization. This vaccine demonstrates broad protection against SARS-CoV-2 variants, with highly attenuated replication and minimal lung pathology across multiple in vivo passages. The vaccine induced robust mucosal and systemic neutralizing antibodies, as well as T-cell responses, in male and female hamsters, female K18-hACE2 mice, and male HFH4-hACE2 mice. In male and female cynomolgus macaques, CDO-7N-1 effectively prevented infection, reduced severe disease, and limited transmission of SARS-CoV-2 variants. This innovative approach offers potential advantages over traditional spike-protein vaccines by providing durable protection and targeting emerging variants to curb virus transmission. Supported by ORIP (K01OD026529).
Characterization of Collaborative Cross Mouse Founder Strain CAST/EiJ as a Novel Model for Lethal COVID-19
Baker et al., Scientific Reports. 2024.
https://www.nature.com/articles/s41598-024-77087-1
Researchers characterized the Collaborative Cross (CC) mouse model founder strain CAST/EiJ as a novel model for severe COVID-19, exhibiting high viral loads and mortality. By leveraging genetically diverse CC strains, this study identified variations in susceptibility and survival against SARS-CoV-2 variants. CAST/EiJ mice developed lung pathology and mortality despite antiviral defenses, making them a valuable tool for understanding host–pathogen interactions. The findings emphasize the utility of diverse animal models in uncovering genetic and immunological factors that influence disease outcomes, facilitating the development of targeted therapies against COVID-19 to mitigate future pandemics. Supported by ORIP (P40OD011102).
Placental Gene Therapy in Nonhuman Primates: A Pilot Study of Maternal, Placental, and Fetal Response to Non-Viral, Polymeric Nanoparticle Delivery of IGF1
Wilson et al., Molecular Human Reproduction. 2024.
https://academic.oup.com/molehr/article/30/11/gaae038/7876288#493719584
This study investigates a novel nanoparticle-mediated gene therapy approach for addressing fetal growth restriction (FGR) in pregnant female nonhuman primates. Using polymer-based nanoparticles delivering a human insulin-like growth factor 1 (IGF1) transgene, the therapy targets the placenta via ultrasound-guided injections. Researchers evaluated maternal, placental, and fetal responses by analyzing tissues, immunomodulatory proteins, and hormones (progesterone and estradiol). Findings highlight the potential of IGF1 nanoparticles to correct placental insufficiency by enhancing fetal growth, providing a groundbreaking advancement for in utero treatments. This research supports further exploration of nonviral gene therapies for improving pregnancy outcomes and combating FGR-related complications. Supported by ORIP (P51OD011106) and NICHD.
Commentary: The International Mouse Phenotyping Consortium: High-Throughput In Vivo Functional Annotation of the Mammalian Genome
Lloyd, Mammalian Genome. 2024.
https://pubmed.ncbi.nlm.nih.gov/39254744
The International Mouse Phenotyping Consortium (IMPC), a collectively governed consortium of 21 academic research institutions across 15 countries on 5 continents, represents a groundbreaking approach in genetics and biomedical research. Its goal is to create a comprehensive catalog of mammalian gene function that is freely available and equally accessible to the global research community. So far, the IMPC has uncovered the function of thousands of genes about which little was previously known. By 2027, when the current round of funding expires, the IMPC will have produced and phenotyped nearly 12,000 knockout mouse lines representing approximately 60% of the human orthologous genome in mice. This new knowledge has produced numerous insights about the role of genes in health and disease, including informing the genetic basis of rare diseases and positing gene product influences on common diseases. However, as IMPC nears the end of the current funding cycle, its path forward remains unclear. Supported by ORIP (UM1OD023221).