Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.

Rett syndrome (RTT) is an autism spectrum disorder (ASD) caused by mutations in the X-linked MECP2 gene that encodes methyl-CpG binding protein 2 (MeCP2). Symptoms range in severity and include psychomotor disabilities, seizures, ataxia, and intellectual disability. Symptom onset is between 6-18 mon...

Full description

Bibliographic Details
Main Authors: Min Jung Park, Susan Aja, Qun Li, Alicia L Degano, Judith Penati, Justin Zhuo, Charles R Roe, Gabriele V Ronnett
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2014-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC4192301?pdf=render
id doaj-892c6d1a265a443aa873bd1712bfba01
record_format Article
spelling doaj-892c6d1a265a443aa873bd1712bfba012020-11-24T22:03:21ZengPublic Library of Science (PLoS)PLoS ONE1932-62032014-01-01910e10952710.1371/journal.pone.0109527Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.Min Jung ParkSusan AjaQun LiAlicia L DeganoJudith PenatiJustin ZhuoCharles R RoeGabriele V RonnettRett syndrome (RTT) is an autism spectrum disorder (ASD) caused by mutations in the X-linked MECP2 gene that encodes methyl-CpG binding protein 2 (MeCP2). Symptoms range in severity and include psychomotor disabilities, seizures, ataxia, and intellectual disability. Symptom onset is between 6-18 months of age, a critical period of brain development that is highly energy-dependent. Notably, patients with RTT have evidence of mitochondrial dysfunction, as well as abnormal levels of the adipokines leptin and adiponectin, suggesting overall metabolic imbalance. We hypothesized that one contributor to RTT symptoms is energy deficiency due to defective nutrient substrate utilization by the TCA cycle. This energy deficit would lead to a metabolic imbalance, but would be treatable by providing anaplerotic substrates to the TCA cycle to enhance energy production. We show that dietary therapy with triheptanoin significantly increased longevity and improved motor function and social interaction in male mice hemizygous for Mecp2 knockout. Anaplerotic therapy in Mecp2 knockout mice also improved indicators of impaired substrate utilization, decreased adiposity, increased glucose tolerance and insulin sensitivity, decreased serum leptin and insulin, and improved mitochondrial morphology in skeletal muscle. Untargeted metabolomics of liver and skeletal muscle revealed increases in levels of TCA cycle intermediates with triheptanoin diet, as well as normalizations of glucose and fatty acid biochemical pathways consistent with the improved metabolic phenotype in Mecp2 knockout mice on triheptanoin. These results suggest that an approach using dietary supplementation with anaplerotic substrate is effective in improving symptoms and metabolic health in RTT.http://europepmc.org/articles/PMC4192301?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Min Jung Park
Susan Aja
Qun Li
Alicia L Degano
Judith Penati
Justin Zhuo
Charles R Roe
Gabriele V Ronnett
spellingShingle Min Jung Park
Susan Aja
Qun Li
Alicia L Degano
Judith Penati
Justin Zhuo
Charles R Roe
Gabriele V Ronnett
Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.
PLoS ONE
author_facet Min Jung Park
Susan Aja
Qun Li
Alicia L Degano
Judith Penati
Justin Zhuo
Charles R Roe
Gabriele V Ronnett
author_sort Min Jung Park
title Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.
title_short Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.
title_full Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.
title_fullStr Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.
title_full_unstemmed Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice.
title_sort anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in mecp2-null mice.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2014-01-01
description Rett syndrome (RTT) is an autism spectrum disorder (ASD) caused by mutations in the X-linked MECP2 gene that encodes methyl-CpG binding protein 2 (MeCP2). Symptoms range in severity and include psychomotor disabilities, seizures, ataxia, and intellectual disability. Symptom onset is between 6-18 months of age, a critical period of brain development that is highly energy-dependent. Notably, patients with RTT have evidence of mitochondrial dysfunction, as well as abnormal levels of the adipokines leptin and adiponectin, suggesting overall metabolic imbalance. We hypothesized that one contributor to RTT symptoms is energy deficiency due to defective nutrient substrate utilization by the TCA cycle. This energy deficit would lead to a metabolic imbalance, but would be treatable by providing anaplerotic substrates to the TCA cycle to enhance energy production. We show that dietary therapy with triheptanoin significantly increased longevity and improved motor function and social interaction in male mice hemizygous for Mecp2 knockout. Anaplerotic therapy in Mecp2 knockout mice also improved indicators of impaired substrate utilization, decreased adiposity, increased glucose tolerance and insulin sensitivity, decreased serum leptin and insulin, and improved mitochondrial morphology in skeletal muscle. Untargeted metabolomics of liver and skeletal muscle revealed increases in levels of TCA cycle intermediates with triheptanoin diet, as well as normalizations of glucose and fatty acid biochemical pathways consistent with the improved metabolic phenotype in Mecp2 knockout mice on triheptanoin. These results suggest that an approach using dietary supplementation with anaplerotic substrate is effective in improving symptoms and metabolic health in RTT.
url http://europepmc.org/articles/PMC4192301?pdf=render
work_keys_str_mv AT minjungpark anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT susanaja anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT qunli anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT alicialdegano anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT judithpenati anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT justinzhuo anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT charlesrroe anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
AT gabrielevronnett anaplerotictriheptanoindietenhancesmitochondrialsubstrateusetoremodelthemetabolomeandimprovelifespanmotorfunctionandsociabilityinmecp2nullmice
_version_ 1725831888370663424