Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging

As the most abundant cell types in the brain, astrocytes form a tissue-wide signaling network that is responsible for maintaining brain homeostasis and regulating various brain activities. Here, we review some of the essential functions that astrocytes perform in supporting neurons, modulating the i...

Full description

Bibliographic Details
Main Authors: Marcela K. Preininger, Daniela Kaufer
Format: Article
Language:English
Published: MDPI AG 2022-06-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/23/11/6217
_version_ 1797493058833481728
author Marcela K. Preininger
Daniela Kaufer
author_facet Marcela K. Preininger
Daniela Kaufer
author_sort Marcela K. Preininger
collection DOAJ
description As the most abundant cell types in the brain, astrocytes form a tissue-wide signaling network that is responsible for maintaining brain homeostasis and regulating various brain activities. Here, we review some of the essential functions that astrocytes perform in supporting neurons, modulating the immune response, and regulating and maintaining the blood–brain barrier (BBB). Given their importance in brain health, it follows that astrocyte dysfunction has detrimental effects. Indeed, dysfunctional astrocytes are implicated in age-related neuropathology and participate in the onset and progression of neurodegenerative diseases. Here, we review two mechanisms by which astrocytes mediate neuropathology in the aging brain. First, age-associated blood–brain barrier dysfunction (BBBD) causes the hyperactivation of TGFβ signaling in astrocytes, which elicits a pro-inflammatory and epileptogenic phenotype. Over time, BBBD-associated astrocyte dysfunction results in hippocampal and cortical neural hyperexcitability and cognitive deficits. Second, senescent astrocytes accumulate in the brain with age and exhibit a decreased functional capacity and the secretion of senescent-associated secretory phenotype (SASP) factors, which contribute to neuroinflammation and neurotoxicity. Both BBBD and senescence progressively increase during aging and are associated with increased risk of neurodegenerative disease, but the relationship between the two has not yet been established. Thus, we discuss the potential relationship between BBBD, TGFβ hyperactivation, and senescence with respect to astrocytes in the context of aging and disease and identify future areas of investigation in the field.
first_indexed 2024-03-10T01:14:33Z
format Article
id doaj.art-ccbb8d73868d4ddc9a8cf5450cfc419e
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-10T01:14:33Z
publishDate 2022-06-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-ccbb8d73868d4ddc9a8cf5450cfc419e2023-11-23T14:11:40ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672022-06-012311621710.3390/ijms23116217Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in AgingMarcela K. Preininger0Daniela Kaufer1Department of Integrative Biology, University of California, Berkeley, CA 94720, USADepartment of Integrative Biology, University of California, Berkeley, CA 94720, USAAs the most abundant cell types in the brain, astrocytes form a tissue-wide signaling network that is responsible for maintaining brain homeostasis and regulating various brain activities. Here, we review some of the essential functions that astrocytes perform in supporting neurons, modulating the immune response, and regulating and maintaining the blood–brain barrier (BBB). Given their importance in brain health, it follows that astrocyte dysfunction has detrimental effects. Indeed, dysfunctional astrocytes are implicated in age-related neuropathology and participate in the onset and progression of neurodegenerative diseases. Here, we review two mechanisms by which astrocytes mediate neuropathology in the aging brain. First, age-associated blood–brain barrier dysfunction (BBBD) causes the hyperactivation of TGFβ signaling in astrocytes, which elicits a pro-inflammatory and epileptogenic phenotype. Over time, BBBD-associated astrocyte dysfunction results in hippocampal and cortical neural hyperexcitability and cognitive deficits. Second, senescent astrocytes accumulate in the brain with age and exhibit a decreased functional capacity and the secretion of senescent-associated secretory phenotype (SASP) factors, which contribute to neuroinflammation and neurotoxicity. Both BBBD and senescence progressively increase during aging and are associated with increased risk of neurodegenerative disease, but the relationship between the two has not yet been established. Thus, we discuss the potential relationship between BBBD, TGFβ hyperactivation, and senescence with respect to astrocytes in the context of aging and disease and identify future areas of investigation in the field.https://www.mdpi.com/1422-0067/23/11/6217astrocytessenescenceblood–brain barrierTGF beta 1albuminneuroinflammation
spellingShingle Marcela K. Preininger
Daniela Kaufer
Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
International Journal of Molecular Sciences
astrocytes
senescence
blood–brain barrier
TGF beta 1
albumin
neuroinflammation
title Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
title_full Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
title_fullStr Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
title_full_unstemmed Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
title_short Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
title_sort blood brain barrier dysfunction and astrocyte senescence as reciprocal drivers of neuropathology in aging
topic astrocytes
senescence
blood–brain barrier
TGF beta 1
albumin
neuroinflammation
url https://www.mdpi.com/1422-0067/23/11/6217
work_keys_str_mv AT marcelakpreininger bloodbrainbarrierdysfunctionandastrocytesenescenceasreciprocaldriversofneuropathologyinaging
AT danielakaufer bloodbrainbarrierdysfunctionandastrocytesenescenceasreciprocaldriversofneuropathologyinaging