Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease. / Salcedo, Claudia; Andersen, Jens V.; Vinten, Kasper Tore; Pinborg, Lars H.; Waagepetersen, Helle S.; Freude, Kristine K.; Aldana, Blanca I.

In: Frontiers in Aging Neuroscience, Vol. 13, 736580, 2021.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Salcedo, C, Andersen, JV, Vinten, KT, Pinborg, LH, Waagepetersen, HS, Freude, KK & Aldana, BI 2021, 'Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease', Frontiers in Aging Neuroscience, vol. 13, 736580. https://doi.org/10.3389/fnagi.2021.736580

APA

Salcedo, C., Andersen, J. V., Vinten, K. T., Pinborg, L. H., Waagepetersen, H. S., Freude, K. K., & Aldana, B. I. (2021). Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease. Frontiers in Aging Neuroscience, 13, [736580]. https://doi.org/10.3389/fnagi.2021.736580

Vancouver

Salcedo C, Andersen JV, Vinten KT, Pinborg LH, Waagepetersen HS, Freude KK et al. Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease. Frontiers in Aging Neuroscience. 2021;13. 736580. https://doi.org/10.3389/fnagi.2021.736580

Author

Salcedo, Claudia ; Andersen, Jens V. ; Vinten, Kasper Tore ; Pinborg, Lars H. ; Waagepetersen, Helle S. ; Freude, Kristine K. ; Aldana, Blanca I. / Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease. In: Frontiers in Aging Neuroscience. 2021 ; Vol. 13.

Bibtex

@article{dab4224e522740ffbd191bf80eff26d0,
title = "Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease",
abstract = "The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are important nitrogen donors for synthesis of glutamate, the main excitatory neurotransmitter in the brain. The glutamate carbon skeleton originates from the tricarboxylic acid (TCA) cycle intermediate α-ketoglutarate, while the amino group is derived from nitrogen donors such as the BCAAs. Disturbances in neurotransmitter homeostasis, mainly of glutamate, are strongly implicated in the pathophysiology of Alzheimer's disease (AD). The divergent BCAA metabolism in different cell types of the human brain is poorly understood, and so is the involvement of astrocytic and neuronal BCAA metabolism in AD. The goal of this study is to provide the first functional characterization of BCAA metabolism in human brain tissue and to investigate BCAA metabolism in AD pathophysiology using astrocytes and neurons derived from human-induced pluripotent stem cells (hiPSCs). Mapping of BCAA metabolism was performed using mass spectrometry and enriched [15N] and [13C] isotopes of leucine, isoleucine, and valine in acutely isolated slices of surgically resected cerebral cortical tissue from human brain and in hiPSC-derived brain cells carrying mutations in either amyloid precursor protein (APP) or presenilin-1 (PSEN-1). We revealed that both human astrocytes of acutely isolated cerebral cortical slices and hiPSC-derived astrocytes were capable of oxidatively metabolizing the carbon skeleton of BCAAs, particularly to support glutamine synthesis. Interestingly, hiPSC-derived astrocytes with APP and PSEN-1 mutations exhibited decreased amino acid synthesis of glutamate, glutamine, and aspartate derived from leucine metabolism. These results clearly demonstrate that there is an active BCAA metabolism in human astrocytes, and that leucine metabolism is selectively impaired in astrocytes derived from the hiPSC models of AD. This impairment in astrocytic BCAA metabolism may contribute to neurotransmitter and energetic imbalances in the AD brain.",
keywords = "AD, astrocytes, BCAA, energy metabolism, glutamate, glutamine, induced pluripotent stem cell, neuron",
author = "Claudia Salcedo and Andersen, {Jens V.} and Vinten, {Kasper Tore} and Pinborg, {Lars H.} and Waagepetersen, {Helle S.} and Freude, {Kristine K.} and Aldana, {Blanca I.}",
note = "Publisher Copyright: {\textcopyright} Copyright {\textcopyright} 2021 Salcedo, Andersen, Vinten, Pinborg, Waagepetersen, Freude and Aldana.",
year = "2021",
doi = "10.3389/fnagi.2021.736580",
language = "English",
volume = "13",
journal = "Frontiers in Aging Neuroscience",
issn = "1663-4365",
publisher = "Frontiers Media S.A.",

}

RIS

TY - JOUR

T1 - Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease

AU - Salcedo, Claudia

AU - Andersen, Jens V.

AU - Vinten, Kasper Tore

AU - Pinborg, Lars H.

AU - Waagepetersen, Helle S.

AU - Freude, Kristine K.

AU - Aldana, Blanca I.

N1 - Publisher Copyright: © Copyright © 2021 Salcedo, Andersen, Vinten, Pinborg, Waagepetersen, Freude and Aldana.

PY - 2021

Y1 - 2021

N2 - The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are important nitrogen donors for synthesis of glutamate, the main excitatory neurotransmitter in the brain. The glutamate carbon skeleton originates from the tricarboxylic acid (TCA) cycle intermediate α-ketoglutarate, while the amino group is derived from nitrogen donors such as the BCAAs. Disturbances in neurotransmitter homeostasis, mainly of glutamate, are strongly implicated in the pathophysiology of Alzheimer's disease (AD). The divergent BCAA metabolism in different cell types of the human brain is poorly understood, and so is the involvement of astrocytic and neuronal BCAA metabolism in AD. The goal of this study is to provide the first functional characterization of BCAA metabolism in human brain tissue and to investigate BCAA metabolism in AD pathophysiology using astrocytes and neurons derived from human-induced pluripotent stem cells (hiPSCs). Mapping of BCAA metabolism was performed using mass spectrometry and enriched [15N] and [13C] isotopes of leucine, isoleucine, and valine in acutely isolated slices of surgically resected cerebral cortical tissue from human brain and in hiPSC-derived brain cells carrying mutations in either amyloid precursor protein (APP) or presenilin-1 (PSEN-1). We revealed that both human astrocytes of acutely isolated cerebral cortical slices and hiPSC-derived astrocytes were capable of oxidatively metabolizing the carbon skeleton of BCAAs, particularly to support glutamine synthesis. Interestingly, hiPSC-derived astrocytes with APP and PSEN-1 mutations exhibited decreased amino acid synthesis of glutamate, glutamine, and aspartate derived from leucine metabolism. These results clearly demonstrate that there is an active BCAA metabolism in human astrocytes, and that leucine metabolism is selectively impaired in astrocytes derived from the hiPSC models of AD. This impairment in astrocytic BCAA metabolism may contribute to neurotransmitter and energetic imbalances in the AD brain.

AB - The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are important nitrogen donors for synthesis of glutamate, the main excitatory neurotransmitter in the brain. The glutamate carbon skeleton originates from the tricarboxylic acid (TCA) cycle intermediate α-ketoglutarate, while the amino group is derived from nitrogen donors such as the BCAAs. Disturbances in neurotransmitter homeostasis, mainly of glutamate, are strongly implicated in the pathophysiology of Alzheimer's disease (AD). The divergent BCAA metabolism in different cell types of the human brain is poorly understood, and so is the involvement of astrocytic and neuronal BCAA metabolism in AD. The goal of this study is to provide the first functional characterization of BCAA metabolism in human brain tissue and to investigate BCAA metabolism in AD pathophysiology using astrocytes and neurons derived from human-induced pluripotent stem cells (hiPSCs). Mapping of BCAA metabolism was performed using mass spectrometry and enriched [15N] and [13C] isotopes of leucine, isoleucine, and valine in acutely isolated slices of surgically resected cerebral cortical tissue from human brain and in hiPSC-derived brain cells carrying mutations in either amyloid precursor protein (APP) or presenilin-1 (PSEN-1). We revealed that both human astrocytes of acutely isolated cerebral cortical slices and hiPSC-derived astrocytes were capable of oxidatively metabolizing the carbon skeleton of BCAAs, particularly to support glutamine synthesis. Interestingly, hiPSC-derived astrocytes with APP and PSEN-1 mutations exhibited decreased amino acid synthesis of glutamate, glutamine, and aspartate derived from leucine metabolism. These results clearly demonstrate that there is an active BCAA metabolism in human astrocytes, and that leucine metabolism is selectively impaired in astrocytes derived from the hiPSC models of AD. This impairment in astrocytic BCAA metabolism may contribute to neurotransmitter and energetic imbalances in the AD brain.

KW - AD

KW - astrocytes

KW - BCAA

KW - energy metabolism

KW - glutamate

KW - glutamine

KW - induced pluripotent stem cell

KW - neuron

U2 - 10.3389/fnagi.2021.736580

DO - 10.3389/fnagi.2021.736580

M3 - Journal article

C2 - 34603012

AN - SCOPUS:85116398881

VL - 13

JO - Frontiers in Aging Neuroscience

JF - Frontiers in Aging Neuroscience

SN - 1663-4365

M1 - 736580

ER -

ID: 282472608