Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Alzheimers Dement ; 20(5): 3455-3471, 2024 May.
Article in English | MEDLINE | ID: mdl-38574388

ABSTRACT

INTRODUCTION: Fundamental questions remain about the key mechanisms that initiate Alzheimer's disease (AD) and the factors that promote its progression. Here we report the successful generation of the first genetically engineered marmosets that carry knock-in (KI) point mutations in the presenilin 1 (PSEN1) gene that can be studied from birth throughout lifespan. METHODS: CRISPR/Cas9 was used to generate marmosets with C410Y or A426P point mutations in PSEN1. Founders and their germline offspring are comprehensively studied longitudinally using non-invasive measures including behavior, biomarkers, neuroimaging, and multiomics signatures. RESULTS: Prior to adulthood, increases in plasma amyloid beta were observed in PSEN1 mutation carriers relative to non-carriers. Analysis of brain revealed alterations in several enzyme-substrate interactions within the gamma secretase complex prior to adulthood. DISCUSSION: Marmosets carrying KI point mutations in PSEN1 provide the opportunity to study the earliest primate-specific mechanisms that contribute to the molecular and cellular root causes of AD onset and progression. HIGHLIGHTS: We report the successful generation of genetically engineered marmosets harboring knock-in point mutations in the PSEN1 gene. PSEN1 marmosets and their germline offspring recapitulate the early emergence of AD-related biomarkers. Studies as early in life as possible in PSEN1 marmosets will enable the identification of primate-specific mechanisms that drive disease progression.


Subject(s)
Alzheimer Disease , Callithrix , Presenilin-1 , Animals , Presenilin-1/genetics , Alzheimer Disease/genetics , Male , Female , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Point Mutation/genetics , Animals, Genetically Modified , CRISPR-Cas Systems , Gene Knock-In Techniques , Mutation/genetics , Humans
2.
Front Immunol ; 11: 891, 2020.
Article in English | MEDLINE | ID: mdl-32477361

ABSTRACT

In 2017 over 550,000 estimated new cases of multi-drug/rifampicin resistant tuberculosis (MDR/RR-TB) occurred, emphasizing a need for new treatment strategies. Linezolid (LZD) is a potent antibiotic for drug-resistant Gram-positive infections and is an effective treatment for TB. However, extended LZD use can lead to LZD-associated host toxicities, most commonly bone marrow suppression. LZD toxicities may be mediated by IL-1, an inflammatory pathway important for early immunity during M. tuberculosis infection. However, IL-1 can contribute to pathology and disease severity late in TB progression. Since IL-1 may contribute to LZD toxicity and does influence TB pathology, we targeted this pathway with a potential host-directed therapy (HDT). We hypothesized LZD efficacy could be enhanced by modulation of IL-1 pathway to reduce bone marrow toxicity and TB associated-inflammation. We used two animal models of TB to test our hypothesis, a TB-susceptible mouse model and clinically relevant cynomolgus macaques. Antagonizing IL-1 in mice with established infection reduced lung neutrophil numbers and partially restored the erythroid progenitor populations that are depleted by LZD. In macaques, we found no conclusive evidence of bone marrow suppression associated with LZD, indicating our treatment time may have been short enough to avoid the toxicities observed in humans. Though treatment was only 4 weeks (the FDA approved regimen at the time of study), we observed sterilization of the majority of granulomas regardless of co-administration of the FDA-approved IL-1 receptor antagonist (IL-1Rn), also known as Anakinra. However, total lung inflammation was significantly reduced in macaques treated with IL-1Rn and LZD compared to LZD alone. Importantly, IL-1Rn administration did not impair the host response against Mtb or LZD efficacy in either animal model. Together, our data support that inhibition of IL-1 in combination with LZD has potential to be an effective HDT for TB and the need for further research in this area.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Linezolid/therapeutic use , Tuberculosis/drug therapy , Animals , Disease Models, Animal , Inflammation , Macaca , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Tuberculosis, Multidrug-Resistant/drug therapy
3.
Eur J Pharm Sci ; 151: 105421, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32531349

ABSTRACT

Non-human primates (NHP) are thought to be a good preclinical animal model for tuberculosis because they develop disease characteristics that are similar to humans. The objective of the current study was to determine if NHPs can also be used to reliably predict the exposure of tedizolid, sutezolid, and its biologically active metabolite sutezolid-M1 in humans. The prodrug tedizolid phosphate and sutezolid were administered orally to NHPs either once or twice daily for up to eight days. The active moieties, tedizolid, and sutezolid showed linear pharmacokinetics and respective concentration-time profiles could be described by one-compartment body models with first-order elimination. One additional metabolite compartment with first-order elimination was found appropriate to capture the pharmacokinetics of sutezolid-M1. Once allometrically scaled to humans with a fixed exponent of 0.75 for apparent clearance and 1 for apparent volume of distribution, the AUCs of tedizolid and sutezolid were predicted reasonably well, whereas Cmax was under-predicted for sutezolid. Both NHP and humanized concentration-time profiles will now be used in vitro hollow-fiber pharmacodynamic experiments to determine if differences in drug exposures result in differences in Mycobacterium tuberculosis kill and emergence of resistance.


Subject(s)
Anti-Bacterial Agents , Oxazoles , Animals , Anti-Bacterial Agents/therapeutic use , Microbial Sensitivity Tests , Organophosphates , Oxazolidinones , Primates , Tetrazoles
4.
J Immunol ; 204(3): 644-659, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31862711

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis, continues to be a major global health problem. Lung granulomas are organized structures of host immune cells that function to contain the bacteria. Cytokine expression is a critical component of the protective immune response, but inappropriate cytokine expression can exacerbate TB. Although the importance of proinflammatory cytokines in controlling M. tuberculosis infection has been established, the effects of anti-inflammatory cytokines, such as IL-10, in TB are less well understood. To investigate the role of IL-10, we used an Ab to neutralize IL-10 in cynomolgus macaques during M. tuberculosis infection. Anti-IL-10-treated nonhuman primates had similar overall disease outcomes compared with untreated control nonhuman primates, but there were immunological changes in granulomas and lymph nodes from anti-IL-10-treated animals. There was less thoracic inflammation and increased cytokine production in lung granulomas and lymph nodes from IL-10-neutralized animals at 3-4 wk postinfection compared with control animals. At 8 wk postinfection, lung granulomas from IL-10-neutralized animals had reduced cytokine production but increased fibrosis relative to control animals. Although these immunological changes did not affect the overall disease burden during the first 8 wk of infection, we paired computational modeling to explore late infection dynamics. Our findings support that early changes occurring in the absence of IL-10 may lead to better bacterial control later during infection. These unique datasets provide insight into the contribution of IL-10 to the immunological balance necessary for granulomas to control bacterial burden and disease pathology in M. tuberculosis infection.


Subject(s)
Granuloma/immunology , Inflammation/immunology , Interleukin-10/metabolism , Lung/pathology , Lymph Nodes/immunology , Mycobacterium tuberculosis/physiology , Tuberculosis/immunology , Animals , Antibodies, Neutralizing/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Immunity , Lung/immunology , Macaca fascicularis , Pulmonary Fibrosis
SELECTION OF CITATIONS
SEARCH DETAIL
...