Your browser doesn't support javascript.
loading
Intermittent Hypoxia Promotes Functional Neuroprotection from Retinal Ischemia in Untreated First-Generation Offspring: Proteomic Mechanistic Insights.
Harman, Jarrod C; Guidry, Jessie J; Gidday, Jeffrey M.
Afiliação
  • Harman JC; Department of Ophthalmology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States.
  • Guidry JJ; Department of Biochemistry and Molecular Biology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States.
  • Gidday JM; Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States.
Invest Ophthalmol Vis Sci ; 61(11): 15, 2020 09 01.
Article em En | MEDLINE | ID: mdl-32910134
ABSTRACT

Purpose:

Stress can lead to short- or long-term changes in phenotype. Accumulating evidence also supports the transmission of maladaptive phenotypes, induced by adverse stressors, through the germline to manifest in subsequent generations, providing a novel mechanistic basis for the heritability of disease. In the present study in mice, we tested the hypothesis that repeated presentations of a nonharmful conditioning stress, demonstrated previously to protect against retinal ischemia, will also provide ischemic protection in the retinae of their untreated, first-generation (F1) adult offspring.

Methods:

Swiss-Webster ND4 outbred mice were mated following a 16-week period of brief, every-other-day conditioning exposures to mild systemic hypoxia (repetitive hypoxic conditioning, RHC). Retinae of their 5-month-old F1 progeny were subjected to unilateral ischemia. Scotopic electroretinography quantified postischemic outcomes. The injury-resilient retinal proteome was revealed by quantitative mass spectrometry, and bioinformatic analyses identified the biochemical pathways and networks in which these differentially expressed proteins operate.

Results:

Significant resilience to injury in both sexes was documented in F1 mice derived from RHC-treated parents, relative to matched F1 adult progeny derived from normoxic control parents. Ischemia-induced increases and decreases in the expression of many visual transduction proteins that are integral to photoreceptor function were abrogated by parental RHC, providing a molecular basis for the observed functional protection.

Conclusions:

Our proteomic analyses provided mechanistic insights into the molecular manifestation of the inherited, injury-resilient phenotype. To our knowledge, this is the first study in a mammalian model documenting the reprogramming of heritability to promote disease resilience in the next generation.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Doenças Retinianas / Vasos Retinianos / Precondicionamento Isquêmico / Proteoma / Proteômica / Neuroproteção / Isquemia Tipo de estudo: Diagnostic_studies / Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Doenças Retinianas / Vasos Retinianos / Precondicionamento Isquêmico / Proteoma / Proteômica / Neuroproteção / Isquemia Tipo de estudo: Diagnostic_studies / Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article