author_facet Brkic, Marjana
Balusu, Sriram
Van Wonterghem, Elien
Gorlé, Nina
Benilova, Iryna
Kremer, Anna
Van Hove, Inge
Moons, Lieve
De Strooper, Bart
Kanazir, Selma
Libert, Claude
Vandenbroucke, Roosmarijn E.
Brkic, Marjana
Balusu, Sriram
Van Wonterghem, Elien
Gorlé, Nina
Benilova, Iryna
Kremer, Anna
Van Hove, Inge
Moons, Lieve
De Strooper, Bart
Kanazir, Selma
Libert, Claude
Vandenbroucke, Roosmarijn E.
author Brkic, Marjana
Balusu, Sriram
Van Wonterghem, Elien
Gorlé, Nina
Benilova, Iryna
Kremer, Anna
Van Hove, Inge
Moons, Lieve
De Strooper, Bart
Kanazir, Selma
Libert, Claude
Vandenbroucke, Roosmarijn E.
spellingShingle Brkic, Marjana
Balusu, Sriram
Van Wonterghem, Elien
Gorlé, Nina
Benilova, Iryna
Kremer, Anna
Van Hove, Inge
Moons, Lieve
De Strooper, Bart
Kanazir, Selma
Libert, Claude
Vandenbroucke, Roosmarijn E.
The Journal of Neuroscience
Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
General Neuroscience
author_sort brkic, marjana
spelling Brkic, Marjana Balusu, Sriram Van Wonterghem, Elien Gorlé, Nina Benilova, Iryna Kremer, Anna Van Hove, Inge Moons, Lieve De Strooper, Bart Kanazir, Selma Libert, Claude Vandenbroucke, Roosmarijn E. 0270-6474 1529-2401 Society for Neuroscience General Neuroscience http://dx.doi.org/10.1523/jneurosci.0006-15.2015 <jats:p>The blood–CSF barrier (BCSFB) consists of a monolayer of choroid plexus epithelial (CPE) cells that maintain CNS homeostasis by producing CSF and restricting the passage of undesirable molecules and pathogens into the brain. Alzheimer's disease is the most common progressive neurodegenerative disorder and is characterized by the presence of amyloid β (Aβ) plaques and neurofibrillary tangles in the brain. Recent research shows that Alzheimer's disease is associated with morphological changes in CPE cells and compromised production of CSF. Here, we studied the direct effects of Aβ on the functionality of the BCSFB. Intracerebroventricular injection of Aβ1–42 oligomers into the cerebral ventricles of mice, a validated Alzheimer's disease model, caused induction of a cascade of detrimental events, including increased inflammatory gene expression in CPE cells and increased levels of proinflammatory cytokines and chemokines in the CSF. It also rapidly affected CPE cell morphology and tight junction protein levels. These changes were associated with loss of BCSFB integrity, as shown by an increase in BCSFB leakage. Aβ1–42 oligomers also increased matrix metalloproteinase (MMP) gene expression in the CPE and its activity in CSF. Interestingly, BCSFB disruption induced by Aβ1–42 oligomers did not occur in the presence of a broad-spectrum MMP inhibitor or in MMP3-deficient mice. These data provide evidence that MMPs are essential for the BCSFB leakage induced by Aβ1–42 oligomers. Our results reveal that Alzheimer's disease-associated soluble Aβ1–42 oligomers induce BCSFB dysfunction and suggest MMPs as a possible therapeutic target.</jats:p><jats:p><jats:bold>SIGNIFICANCE STATEMENT</jats:bold>No treatments are yet available to cure Alzheimer's disease; however, soluble Aβ oligomers are believed to play a crucial role in the neuroinflammation that is observed in this disease. Here, we studied the effect of Aβ oligomers on the often neglected barrier between blood and brain, called the blood–CSF barrier (BCSFB). This BCSFB is formed by the choroid plexus epithelial cells and is important in maintaining brain homeostasis. We observed Aβ oligomer-induced changes in morphology and loss of BCSFB integrity that might play a role in Alzheimer's disease progression. Strikingly, both inhibition of matrix metalloproteinase (MMP) activity and MMP3 deficiency could protect against the detrimental effects of Aβ oligomer. Clearly, our results suggest that MMP inhibition might have therapeutic potential.</jats:p> Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases The Journal of Neuroscience
doi_str_mv 10.1523/jneurosci.0006-15.2015
facet_avail Online
Free
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTUyMy9qbmV1cm9zY2kuMDAwNi0xNS4yMDE1
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTUyMy9qbmV1cm9zY2kuMDAwNi0xNS4yMDE1
institution DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Rs1
DE-Pl11
DE-105
DE-14
DE-Ch1
DE-L229
imprint Society for Neuroscience, 2015
imprint_str_mv Society for Neuroscience, 2015
issn 0270-6474
1529-2401
issn_str_mv 0270-6474
1529-2401
language English
mega_collection Society for Neuroscience (CrossRef)
match_str brkic2015amyloidboligomersdisruptbloodcsfbarrierintegritybyactivatingmatrixmetalloproteinases
publishDateSort 2015
publisher Society for Neuroscience
recordtype ai
record_format ai
series The Journal of Neuroscience
source_id 49
title Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_unstemmed Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_full Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_fullStr Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_full_unstemmed Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_short Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_sort amyloid β oligomers disrupt blood–csf barrier integrity by activating matrix metalloproteinases
topic General Neuroscience
url http://dx.doi.org/10.1523/jneurosci.0006-15.2015
publishDate 2015
physical 12766-12778
description <jats:p>The blood–CSF barrier (BCSFB) consists of a monolayer of choroid plexus epithelial (CPE) cells that maintain CNS homeostasis by producing CSF and restricting the passage of undesirable molecules and pathogens into the brain. Alzheimer's disease is the most common progressive neurodegenerative disorder and is characterized by the presence of amyloid β (Aβ) plaques and neurofibrillary tangles in the brain. Recent research shows that Alzheimer's disease is associated with morphological changes in CPE cells and compromised production of CSF. Here, we studied the direct effects of Aβ on the functionality of the BCSFB. Intracerebroventricular injection of Aβ1–42 oligomers into the cerebral ventricles of mice, a validated Alzheimer's disease model, caused induction of a cascade of detrimental events, including increased inflammatory gene expression in CPE cells and increased levels of proinflammatory cytokines and chemokines in the CSF. It also rapidly affected CPE cell morphology and tight junction protein levels. These changes were associated with loss of BCSFB integrity, as shown by an increase in BCSFB leakage. Aβ1–42 oligomers also increased matrix metalloproteinase (MMP) gene expression in the CPE and its activity in CSF. Interestingly, BCSFB disruption induced by Aβ1–42 oligomers did not occur in the presence of a broad-spectrum MMP inhibitor or in MMP3-deficient mice. These data provide evidence that MMPs are essential for the BCSFB leakage induced by Aβ1–42 oligomers. Our results reveal that Alzheimer's disease-associated soluble Aβ1–42 oligomers induce BCSFB dysfunction and suggest MMPs as a possible therapeutic target.</jats:p><jats:p><jats:bold>SIGNIFICANCE STATEMENT</jats:bold>No treatments are yet available to cure Alzheimer's disease; however, soluble Aβ oligomers are believed to play a crucial role in the neuroinflammation that is observed in this disease. Here, we studied the effect of Aβ oligomers on the often neglected barrier between blood and brain, called the blood–CSF barrier (BCSFB). This BCSFB is formed by the choroid plexus epithelial cells and is important in maintaining brain homeostasis. We observed Aβ oligomer-induced changes in morphology and loss of BCSFB integrity that might play a role in Alzheimer's disease progression. Strikingly, both inhibition of matrix metalloproteinase (MMP) activity and MMP3 deficiency could protect against the detrimental effects of Aβ oligomer. Clearly, our results suggest that MMP inhibition might have therapeutic potential.</jats:p>
container_issue 37
container_start_page 12766
container_title The Journal of Neuroscience
container_volume 35
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792346266568491008
geogr_code not assigned
last_indexed 2024-03-01T17:36:39.635Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Amyloid+%CE%B2+Oligomers+Disrupt+Blood%E2%80%93CSF+Barrier+Integrity+by+Activating+Matrix+Metalloproteinases&rft.date=2015-09-16&genre=article&issn=1529-2401&volume=35&issue=37&spage=12766&epage=12778&pages=12766-12778&jtitle=The+Journal+of+Neuroscience&atitle=Amyloid+%CE%B2+Oligomers+Disrupt+Blood%E2%80%93CSF+Barrier+Integrity+by+Activating+Matrix+Metalloproteinases&aulast=Vandenbroucke&aufirst=Roosmarijn+E.&rft_id=info%3Adoi%2F10.1523%2Fjneurosci.0006-15.2015&rft.language%5B0%5D=eng
SOLR
_version_ 1792346266568491008
author Brkic, Marjana, Balusu, Sriram, Van Wonterghem, Elien, Gorlé, Nina, Benilova, Iryna, Kremer, Anna, Van Hove, Inge, Moons, Lieve, De Strooper, Bart, Kanazir, Selma, Libert, Claude, Vandenbroucke, Roosmarijn E.
author_facet Brkic, Marjana, Balusu, Sriram, Van Wonterghem, Elien, Gorlé, Nina, Benilova, Iryna, Kremer, Anna, Van Hove, Inge, Moons, Lieve, De Strooper, Bart, Kanazir, Selma, Libert, Claude, Vandenbroucke, Roosmarijn E., Brkic, Marjana, Balusu, Sriram, Van Wonterghem, Elien, Gorlé, Nina, Benilova, Iryna, Kremer, Anna, Van Hove, Inge, Moons, Lieve, De Strooper, Bart, Kanazir, Selma, Libert, Claude, Vandenbroucke, Roosmarijn E.
author_sort brkic, marjana
container_issue 37
container_start_page 12766
container_title The Journal of Neuroscience
container_volume 35
description <jats:p>The blood–CSF barrier (BCSFB) consists of a monolayer of choroid plexus epithelial (CPE) cells that maintain CNS homeostasis by producing CSF and restricting the passage of undesirable molecules and pathogens into the brain. Alzheimer's disease is the most common progressive neurodegenerative disorder and is characterized by the presence of amyloid β (Aβ) plaques and neurofibrillary tangles in the brain. Recent research shows that Alzheimer's disease is associated with morphological changes in CPE cells and compromised production of CSF. Here, we studied the direct effects of Aβ on the functionality of the BCSFB. Intracerebroventricular injection of Aβ1–42 oligomers into the cerebral ventricles of mice, a validated Alzheimer's disease model, caused induction of a cascade of detrimental events, including increased inflammatory gene expression in CPE cells and increased levels of proinflammatory cytokines and chemokines in the CSF. It also rapidly affected CPE cell morphology and tight junction protein levels. These changes were associated with loss of BCSFB integrity, as shown by an increase in BCSFB leakage. Aβ1–42 oligomers also increased matrix metalloproteinase (MMP) gene expression in the CPE and its activity in CSF. Interestingly, BCSFB disruption induced by Aβ1–42 oligomers did not occur in the presence of a broad-spectrum MMP inhibitor or in MMP3-deficient mice. These data provide evidence that MMPs are essential for the BCSFB leakage induced by Aβ1–42 oligomers. Our results reveal that Alzheimer's disease-associated soluble Aβ1–42 oligomers induce BCSFB dysfunction and suggest MMPs as a possible therapeutic target.</jats:p><jats:p><jats:bold>SIGNIFICANCE STATEMENT</jats:bold>No treatments are yet available to cure Alzheimer's disease; however, soluble Aβ oligomers are believed to play a crucial role in the neuroinflammation that is observed in this disease. Here, we studied the effect of Aβ oligomers on the often neglected barrier between blood and brain, called the blood–CSF barrier (BCSFB). This BCSFB is formed by the choroid plexus epithelial cells and is important in maintaining brain homeostasis. We observed Aβ oligomer-induced changes in morphology and loss of BCSFB integrity that might play a role in Alzheimer's disease progression. Strikingly, both inhibition of matrix metalloproteinase (MMP) activity and MMP3 deficiency could protect against the detrimental effects of Aβ oligomer. Clearly, our results suggest that MMP inhibition might have therapeutic potential.</jats:p>
doi_str_mv 10.1523/jneurosci.0006-15.2015
facet_avail Online, Free
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTUyMy9qbmV1cm9zY2kuMDAwNi0xNS4yMDE1
imprint Society for Neuroscience, 2015
imprint_str_mv Society for Neuroscience, 2015
institution DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Rs1, DE-Pl11, DE-105, DE-14, DE-Ch1, DE-L229
issn 0270-6474, 1529-2401
issn_str_mv 0270-6474, 1529-2401
language English
last_indexed 2024-03-01T17:36:39.635Z
match_str brkic2015amyloidboligomersdisruptbloodcsfbarrierintegritybyactivatingmatrixmetalloproteinases
mega_collection Society for Neuroscience (CrossRef)
physical 12766-12778
publishDate 2015
publishDateSort 2015
publisher Society for Neuroscience
record_format ai
recordtype ai
series The Journal of Neuroscience
source_id 49
spelling Brkic, Marjana Balusu, Sriram Van Wonterghem, Elien Gorlé, Nina Benilova, Iryna Kremer, Anna Van Hove, Inge Moons, Lieve De Strooper, Bart Kanazir, Selma Libert, Claude Vandenbroucke, Roosmarijn E. 0270-6474 1529-2401 Society for Neuroscience General Neuroscience http://dx.doi.org/10.1523/jneurosci.0006-15.2015 <jats:p>The blood–CSF barrier (BCSFB) consists of a monolayer of choroid plexus epithelial (CPE) cells that maintain CNS homeostasis by producing CSF and restricting the passage of undesirable molecules and pathogens into the brain. Alzheimer's disease is the most common progressive neurodegenerative disorder and is characterized by the presence of amyloid β (Aβ) plaques and neurofibrillary tangles in the brain. Recent research shows that Alzheimer's disease is associated with morphological changes in CPE cells and compromised production of CSF. Here, we studied the direct effects of Aβ on the functionality of the BCSFB. Intracerebroventricular injection of Aβ1–42 oligomers into the cerebral ventricles of mice, a validated Alzheimer's disease model, caused induction of a cascade of detrimental events, including increased inflammatory gene expression in CPE cells and increased levels of proinflammatory cytokines and chemokines in the CSF. It also rapidly affected CPE cell morphology and tight junction protein levels. These changes were associated with loss of BCSFB integrity, as shown by an increase in BCSFB leakage. Aβ1–42 oligomers also increased matrix metalloproteinase (MMP) gene expression in the CPE and its activity in CSF. Interestingly, BCSFB disruption induced by Aβ1–42 oligomers did not occur in the presence of a broad-spectrum MMP inhibitor or in MMP3-deficient mice. These data provide evidence that MMPs are essential for the BCSFB leakage induced by Aβ1–42 oligomers. Our results reveal that Alzheimer's disease-associated soluble Aβ1–42 oligomers induce BCSFB dysfunction and suggest MMPs as a possible therapeutic target.</jats:p><jats:p><jats:bold>SIGNIFICANCE STATEMENT</jats:bold>No treatments are yet available to cure Alzheimer's disease; however, soluble Aβ oligomers are believed to play a crucial role in the neuroinflammation that is observed in this disease. Here, we studied the effect of Aβ oligomers on the often neglected barrier between blood and brain, called the blood–CSF barrier (BCSFB). This BCSFB is formed by the choroid plexus epithelial cells and is important in maintaining brain homeostasis. We observed Aβ oligomer-induced changes in morphology and loss of BCSFB integrity that might play a role in Alzheimer's disease progression. Strikingly, both inhibition of matrix metalloproteinase (MMP) activity and MMP3 deficiency could protect against the detrimental effects of Aβ oligomer. Clearly, our results suggest that MMP inhibition might have therapeutic potential.</jats:p> Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases The Journal of Neuroscience
spellingShingle Brkic, Marjana, Balusu, Sriram, Van Wonterghem, Elien, Gorlé, Nina, Benilova, Iryna, Kremer, Anna, Van Hove, Inge, Moons, Lieve, De Strooper, Bart, Kanazir, Selma, Libert, Claude, Vandenbroucke, Roosmarijn E., The Journal of Neuroscience, Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases, General Neuroscience
title Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_full Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_fullStr Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_full_unstemmed Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_short Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
title_sort amyloid β oligomers disrupt blood–csf barrier integrity by activating matrix metalloproteinases
title_unstemmed Amyloid β Oligomers Disrupt Blood–CSF Barrier Integrity by Activating Matrix Metalloproteinases
topic General Neuroscience
url http://dx.doi.org/10.1523/jneurosci.0006-15.2015