1
|
Aplin C, Zielinski KA, Pabit S, Ogunribido D, Katt WP, Pollack L, Cerione RA, Milano SK. Distinct conformational states enable transglutaminase 2 to promote cancer cell survival versus cell death. Commun Biol 2024; 7:982. [PMID: 39134806 PMCID: PMC11319651 DOI: 10.1038/s42003-024-06672-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Transglutaminase 2 (TG2) is a GTP-binding, protein-crosslinking enzyme that has been investigated as a therapeutic target for Celiac disease, neurological disorders, and aggressive cancers. TG2 has been suggested to adopt two conformational states that regulate its functions: a GTP-bound, closed conformation, and a calcium-bound, crosslinking-active open conformation. TG2 mutants that constitutively adopt an open conformation are cytotoxic to cancer cells. Thus, small molecules that bind and stabilize the open conformation of TG2 could offer a new therapeutic strategy. Here, we investigate TG2, using static and time-resolved small-angle X-ray scattering (SAXS) and single-particle cryoelectron microscopy (cryo-EM), to determine the conformational states responsible for conferring its biological effects. We also describe a newly developed TG2 inhibitor, LM11, that potently kills glioblastoma cells and use SAXS to investigate how LM11 affects the conformational states of TG2. Using SAXS and cryo-EM, we show that guanine nucleotides bind and stabilize a monomeric closed conformation while calcium binds to an open state that can form higher order oligomers. SAXS analysis suggests how a TG2 mutant that constitutively adopts the open state binds nucleotides through an alternative mechanism to wildtype TG2. Furthermore, we use time resolved SAXS to show that LM11 increases the ability of calcium to bind and stabilize an open conformation, which is not reversible by guanine nucleotides and is cytotoxic to cancer cells. Taken together, our findings demonstrate that the conformational dynamics of TG2 are more complex than previously suggested and highlight how conformational stabilization of TG2 by LM11 maintains TG2 in a cytotoxic conformational state.
Collapse
Affiliation(s)
- Cody Aplin
- Department of Chemistry and Chemical Biology, Cornell University, 14853, Ithaca, NY, USA
- Department of Molecular Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Kara A Zielinski
- School of Applied and Engineering Physics, Cornell University, 14853, Ithaca, NY, USA
| | - Suzette Pabit
- School of Applied and Engineering Physics, Cornell University, 14853, Ithaca, NY, USA
| | - Deborah Ogunribido
- Department of Chemistry and Chemical Biology, Cornell University, 14853, Ithaca, NY, USA
| | - William P Katt
- Department of Molecular Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Lois Pollack
- School of Applied and Engineering Physics, Cornell University, 14853, Ithaca, NY, USA
| | - Richard A Cerione
- Department of Chemistry and Chemical Biology, Cornell University, 14853, Ithaca, NY, USA.
- Department of Molecular Medicine, Cornell University, 14853, Ithaca, NY, USA.
| | - Shawn K Milano
- Department of Chemistry and Chemical Biology, Cornell University, 14853, Ithaca, NY, USA
- Department of Molecular Medicine, Cornell University, 14853, Ithaca, NY, USA
| |
Collapse
|
2
|
Xiong W, Cai J, Sun B, Lin H, Wei C, Huang C, Zhu X, Tan H. The association between genetic variations and morphology-based brain networks changes in Alzheimer's disease. J Neurochem 2024; 168:1490-1502. [PMID: 36625269 DOI: 10.1111/jnc.15761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/18/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is a highly heritable disease. The morphological changes of cortical cortex (such as, cortical thickness and surface area) in AD always accompany by the change of the functional connectivity to other brain regions and influence the short- and long-range brain network connections, causing functional deficits of AD. In this study, the first hypothesis is that genetic variations might affect morphology-based brain networks, leading to functional deficits; the second hypothesis is that protein-protein interaction (PPI) between the candidate proteins and known interacting proteins to AD might exist and influence AD. 600 470 variants and structural magnetic resonance imaging scans from 175 AD patients and 214 healthy controls were obtained from the Alzheimer's Disease Neuroimaging Initiative-1 database. A co-sparse reduced-rank regression model was fit to study the relationship between non-synonymous mutations and morphology-based brain networks. After that, PPIs between selected genes and BACE1, an enzyme that was known to be related to AD, are explored by using molecular dynamics (MD) simulation and co-immunoprecipitation (Co-IP) experiments. Eight genes affecting morphology-based brain networks were identified. The results of MD simulation showed that the PPI between TGM4 and BACE1 was the strongest among them and its interaction was verified by Co-IP. Hence, gene variations influence morphology-based brain networks in AD, leading to functional deficits. This finding, validated by MD simulation and Co-IP, suggests that the effect is robust.
Collapse
Affiliation(s)
- Weixue Xiong
- Shantou University Medical College, Shantou, China
| | - Jiahui Cai
- Shantou University Medical College, Shantou, China
| | - Bo Sun
- Department of Radiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Henghui Lin
- Shantou University Medical College, Shantou, China
| | - Chiyu Wei
- Shantou University Medical College, Shantou, China
| | | | - Xiaohui Zhu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Haizhu Tan
- Shantou University Medical College, Shantou, China
| |
Collapse
|
3
|
Liu J, Mouradian MM. Pathogenetic Contributions and Therapeutic Implications of Transglutaminase 2 in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2364. [PMID: 38397040 PMCID: PMC10888553 DOI: 10.3390/ijms25042364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases encompass a heterogeneous group of disorders that afflict millions of people worldwide. Characteristic protein aggregates are histopathological hallmark features of these disorders, including Amyloid β (Aβ)-containing plaques and tau-containing neurofibrillary tangles in Alzheimer's disease, α-Synuclein (α-Syn)-containing Lewy bodies and Lewy neurites in Parkinson's disease and dementia with Lewy bodies, and mutant huntingtin (mHTT) in nuclear inclusions in Huntington's disease. These various aggregates are found in specific brain regions that are impacted by neurodegeneration and associated with clinical manifestations. Transglutaminase (TG2) (also known as tissue transglutaminase) is the most ubiquitously expressed member of the transglutaminase family with protein crosslinking activity. To date, Aβ, tau, α-Syn, and mHTT have been determined to be substrates of TG2, leading to their aggregation and implicating the involvement of TG2 in several pathophysiological events in neurodegenerative disorders. In this review, we summarize the biochemistry and physiologic functions of TG2 and describe recent advances in the pathogenetic role of TG2 in these diseases. We also review TG2 inhibitors tested in clinical trials and discuss recent TG2-targeting approaches, which offer new perspectives for the design of future highly potent and selective drugs with improved brain delivery as a disease-modifying treatment for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
4
|
Aplin C, Zielinski KA, Pabit S, Ogunribido D, Katt WP, Pollack L, Cerione RA, Milano SK. Defining the conformational states that enable transglutaminase 2 to promote cancer cell survival versus cell death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.04.578794. [PMID: 38370687 PMCID: PMC10871292 DOI: 10.1101/2024.02.04.578794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Transglutaminase 2 (TG2) is a GTP-binding/protein-crosslinking enzyme that has been investigated as a therapeutic target for Celiac disease, neurological disorders, and aggressive cancers. TG2 has been suggested to adopt two conformational states that regulate its functions: a GTP-bound, closed conformation, and a calcium-bound, crosslinking-active open conformation. TG2 mutants that constitutively adopt an open conformation are cytotoxic to cancer cells. Thus, small molecules that maintain the open conformation of TG2 could offer a new therapeutic strategy. Here, we investigate TG2, using static and time-resolved small-angle X-ray scattering (SAXS) and single-particle cryoelectron microscopy (cryo-EM), to determine the conformational states responsible for conferring its biological effects. We also describe a newly developed TG2 inhibitor, LM11, that potently kills glioblastoma cells and use SAXS to investigate how LM11 affects the conformational states of TG2. Using SAXS and cryo-EM, we show that guanine nucleotide-bound TG2 adopts a monomeric closed conformation while calcium-bound TG2 assumes an open conformational state that can form higher order oligomers. SAXS analysis also suggests how a TG2 mutant that constitutively adopts the open state binds nucleotides through an alternative mechanism to wildtype TG2. Furthermore, we use time-resolved SAXS to show that LM11 increases the ability of calcium to drive TG2 to an open conformation, which is not reversible by guanine nucleotides and is cytotoxic to cancer cells. Taken together, our findings demonstrate that the conformational dynamics of TG2 are more complex than previously suggested and highlight how conformational stabilization of TG2 by LM11 maintains TG2 in a cytotoxic conformational state.
Collapse
Affiliation(s)
- Cody Aplin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Kara A. Zielinski
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853
| | - Suzette Pabit
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853
| | - Deborah Ogunribido
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - William P. Katt
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Lois Pollack
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853
| | - Richard A. Cerione
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Shawn K. Milano
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
5
|
Handa T, Sasaki H, Takao M, Tano M, Uchida Y. Proteomics-based investigation of cerebrovascular molecular mechanisms in cerebral amyloid angiopathy by the FFPE-LMD-PCT-SWATH method. Fluids Barriers CNS 2022; 19:56. [PMID: 35778717 PMCID: PMC9250250 DOI: 10.1186/s12987-022-00351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) occurs in 80% of patients with Alzheimer’s disease (AD) and is mainly caused by the abnormal deposition of Aβ in the walls of cerebral blood vessels. Cerebrovascular molecular mechanisms in CAA were investigated by using comprehensive and accurate quantitative proteomics. Methods Concerning the molecular mechanisms specific to CAA, formalin-fixed paraffin-embedded (FFPE) sections were prepared from patients having AD neuropathologic change (ADNC) with severe cortical Aβ vascular deposition (ADNC +/CAA +), and from patients having ADNC without vascular deposition of Aβ (ADNC +/CAA −; so called, AD). Cerebral cortical vessels were isolated from FFPE sections using laser microdissection (LMD), processed by pressure cycling technology (PCT), and applied to SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Results The protein expression levels of 17 proteins in ADNC +/CAA +/H donors (ADNC +/CAA + donors with highly abundant Aβ in capillaries) were significantly different from those in ADNC +/CAA − and ADNC −/CAA − donors. Furthermore, we identified 56 proteins showing more than a 1.5-fold difference in average expression levels between ADNC +/CAA + and ADNC −/CAA − donors, and were significantly correlated with the levels of Aβ or Collagen alpha-2(VI) chain (COL6A2) (CAA markers) in 11 donors (6 ADNC +/CAA + and 5 ADNC −/CAA −). Over 70% of the 56 proteins showed ADNC +/CAA + specific changes in protein expression. The comparative analysis with brain parenchyma showed that more than 90% of the 56 proteins were vascular-specific pathological changes. A literature-based pathway analysis showed that 42 proteins are associated with fibrosis, oxidative stress and apoptosis. This included the increased expression of Heat shock protein HSP 90-alpha, CD44 antigen and Carbonic anhydrase 1 which are inhibited by potential drugs against CAA. Conclusions The combination of LMD-based isolation of vessels from FFPE sections, PCT-assisted sample processing and SWATH analysis (FFPE-LMD-PCT-SWATH method) revealed for the first time the changes in the expression of many proteins that are involved in fibrosis, ROS production and cell death in ADNC +/CAA + (CAA patients) vessels. The findings reported herein would be useful for developing a better understanding of the pathology of CAA and for promoting the discovery and development of drugs and biomarkers for CAA. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00351-x.
Collapse
Affiliation(s)
- Takumi Handa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hayate Sasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan.,Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan. .,Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
6
|
Wilhelmus MMM, Chouchane O, Loos M, Jongenelen CAM, Brevé JJP, Jonker A, Bol JGJM, Smit AB, Drukarch B. Absence of tissue transglutaminase reduces amyloid-beta pathology in APP23 mice. Neuropathol Appl Neurobiol 2022; 48:e12796. [PMID: 35141929 PMCID: PMC9304226 DOI: 10.1111/nan.12796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/11/2022] [Accepted: 02/05/2022] [Indexed: 11/29/2022]
Abstract
Aims Alzheimer's disease (AD) is characterised by amyloid‐beta (Aβ) aggregates in the brain. Targeting Aβ aggregates is a major approach for AD therapies, although attempts have had little to no success so far. A novel treatment option is to focus on blocking the actual formation of Aβ multimers. The enzyme tissue transglutaminase (TG2) is abundantly expressed in the human brain and plays a key role in post‐translational modifications in Aβ resulting in covalently cross‐linked, stable and neurotoxic Aβ oligomers. In vivo absence of TG2 in the APP23 mouse model may provide evidence that TG2 plays a key role in development and/or progression of Aβ‐related pathology. Methods Here, we compared the effects on Aβ pathology in the presence or absence of TG2 using 12‐month‐old wild type, APP23 and a crossbreed of the TG2−/− mouse model and APP23 mice (APP23/TG2−/−). Results Using immunohistochemistry, we found that the number of Aβ deposits was significantly reduced in the absence of TG2 compared with age‐matched APP23 mice. To pinpoint possible TG2‐associated mechanisms involved in this observation, we analysed soluble brain Aβ1–40, Aβ1–42 and/or Aβ40/42 ratio, and mRNA levels of human APP and TG2 family members present in brain of the various mouse models. In addition, using immunohistochemistry, both beta‐pleated sheet formation in Aβ deposits and the presence of reactive astrocytes associated with Aβ deposits were analysed. Conclusions We found that absence of TG2 reduces the formation of Aβ pathology in the APP23 mouse model, suggesting that TG2 may be a suitable therapeutic target for reducing Aβ deposition in AD.
Collapse
Affiliation(s)
- Micha M M Wilhelmus
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Osoul Chouchane
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Maarten Loos
- Sylics (Synaptologics BV), Amsterdam, The Netherlands
| | - Cornelis A M Jongenelen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - John J P Brevé
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Allert Jonker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - John G J M Bol
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, VU University Amsterdam, The Netherlands
| | - Benjamin Drukarch
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Wilhelmus MMM, Tonoli E, Coveney C, Boocock DJ, Jongenelen CAM, Brevé JJP, Verderio EAM, Drukarch B. The Transglutaminase-2 Interactome in the APP23 Mouse Model of Alzheimer's Disease. Cells 2022; 11:cells11030389. [PMID: 35159198 PMCID: PMC8834516 DOI: 10.3390/cells11030389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/06/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
Amyloid-beta (Aβ) deposition in the brain is closely linked with the development of Alzheimer’s disease (AD). Unfortunately, therapies specifically targeting Aβ deposition have failed to reach their primary clinical endpoints, emphasizing the need to broaden the search strategy for alternative targets/mechanisms. Transglutaminase-2 (TG2) catalyzes post-translational modifications, is present in AD lesions and interacts with AD-associated proteins. However, an unbiased overview of TG2 interactors is lacking in both control and AD brain. Here we aimed to identify these interactors using a crossbreed of the AD-mimicking APP23 mouse model with wild type and TG2 knock-out (TG2−/−) mice. We found that absence of TG2 had no (statistically) significant effect on Aβ pathology, soluble brain levels of Aβ1–40 and Aβ1–42, and mRNA levels of TG family members compared to APP23 mice at 18 months of age. Quantitative proteomics and network analysis revealed a large cluster of TG2 interactors involved in synaptic transmission/assembly and cell adhesion in the APP23 brain typical of AD. Comparative proteomics of wild type and TG2−/− brains revealed a TG2-linked pathological proteome consistent with alterations in both pathways. Our data show that TG2 deletion leads to considerable network alterations consistent with a TG2 role in (dys)regulation of synaptic transmission and cell adhesion in APP23 brains.
Collapse
Affiliation(s)
- Micha M. M. Wilhelmus
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (M.M.M.W.); (C.A.M.J.); (J.J.P.B.); (B.D.)
| | - Elisa Tonoli
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK; (E.T.); (C.C.); (D.J.B.)
| | - Clare Coveney
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK; (E.T.); (C.C.); (D.J.B.)
| | - David J. Boocock
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK; (E.T.); (C.C.); (D.J.B.)
| | - Cornelis A. M. Jongenelen
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (M.M.M.W.); (C.A.M.J.); (J.J.P.B.); (B.D.)
| | - John J. P. Brevé
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (M.M.M.W.); (C.A.M.J.); (J.J.P.B.); (B.D.)
| | - Elisabetta A. M. Verderio
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK; (E.T.); (C.C.); (D.J.B.)
- Department of Biological Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
- Correspondence: ; Tel.: +44-115-8486628
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (M.M.M.W.); (C.A.M.J.); (J.J.P.B.); (B.D.)
| |
Collapse
|
8
|
Lecordier S, Manrique-Castano D, El Moghrabi Y, ElAli A. Neurovascular Alterations in Vascular Dementia: Emphasis on Risk Factors. Front Aging Neurosci 2021; 13:727590. [PMID: 34566627 PMCID: PMC8461067 DOI: 10.3389/fnagi.2021.727590] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular dementia (VaD) constitutes the second most prevalent cause of dementia in the world after Alzheimer’s disease (AD). VaD regroups heterogeneous neurological conditions in which the decline of cognitive functions, including executive functions, is associated with structural and functional alterations in the cerebral vasculature. Among these cerebrovascular disorders, major stroke, and cerebral small vessel disease (cSVD) constitute the major risk factors for VaD. These conditions alter neurovascular functions leading to blood-brain barrier (BBB) deregulation, neurovascular coupling dysfunction, and inflammation. Accumulation of neurovascular impairments over time underlies the cognitive function decline associated with VaD. Furthermore, several vascular risk factors, such as hypertension, obesity, and diabetes have been shown to exacerbate neurovascular impairments and thus increase VaD prevalence. Importantly, air pollution constitutes an underestimated risk factor that triggers vascular dysfunction via inflammation and oxidative stress. The review summarizes the current knowledge related to the pathological mechanisms linking neurovascular impairments associated with stroke, cSVD, and vascular risk factors with a particular emphasis on air pollution, to VaD etiology and progression. Furthermore, the review discusses the major challenges to fully elucidate the pathobiology of VaD, as well as research directions to outline new therapeutic interventions.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel Manrique-Castano
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Yara El Moghrabi
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
9
|
Viejo L, Noori A, Merrill E, Das S, Hyman BT, Serrano-Pozo A. Systematic review of human post-mortem immunohistochemical studies and bioinformatics analyses unveil the complexity of astrocyte reaction in Alzheimer's disease. Neuropathol Appl Neurobiol 2021; 48:e12753. [PMID: 34297416 PMCID: PMC8766893 DOI: 10.1111/nan.12753] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
AIMS Reactive astrocytes in Alzheimer's disease (AD) have traditionally been demonstrated by increased glial fibrillary acidic protein (GFAP) immunoreactivity; however, astrocyte reaction is a complex and heterogeneous phenomenon involving multiple astrocyte functions beyond cytoskeletal remodelling. To better understand astrocyte reaction in AD, we conducted a systematic review of astrocyte immunohistochemical studies in post-mortem AD brains followed by bioinformatics analyses on the extracted reactive astrocyte markers. METHODS NCBI PubMed, APA PsycInfo and WoS-SCIE databases were interrogated for original English research articles with the search terms 'Alzheimer's disease' AND 'astrocytes.' Bioinformatics analyses included protein-protein interaction network analysis, pathway enrichment, and transcription factor enrichment, as well as comparison with public human -omics datasets. RESULTS A total of 306 articles meeting eligibility criteria rendered 196 proteins, most of which were reported to be upregulated in AD vs control brains. Besides cytoskeletal remodelling (e.g., GFAP), bioinformatics analyses revealed a wide range of functional alterations including neuroinflammation (e.g., IL6, MAPK1/3/8 and TNF), oxidative stress and antioxidant defence (e.g., MT1A/2A, NFE2L2, NOS1/2/3, PRDX6 and SOD1/2), lipid metabolism (e.g., APOE, CLU and LRP1), proteostasis (e.g., cathepsins, CRYAB and HSPB1/2/6/8), extracellular matrix organisation (e.g., CD44, MMP1/3 and SERPINA3), and neurotransmission (e.g., CHRNA7, GABA, GLUL, GRM5, MAOB and SLC1A2), among others. CTCF and ESR1 emerged as potential transcription factors driving these changes. Comparison with published -omics datasets validated our results, demonstrating a significant overlap with reported transcriptomic and proteomic changes in AD brains and/or CSF. CONCLUSIONS Our systematic review of the neuropathological literature reveals the complexity of AD reactive astrogliosis. We have shared these findings as an online resource available at www.astrocyteatlas.org.
Collapse
Affiliation(s)
- Lucía Viejo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ayush Noori
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Harvard College, Cambridge, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA
| | - Emily Merrill
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
10
|
Young KZ, Xu G, Keep SG, Borjigin J, Wang MM. Overlapping Protein Accumulation Profiles of CADASIL and CAA: Is There a Common Mechanism Driving Cerebral Small-Vessel Disease? THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:1871-1887. [PMID: 33387456 DOI: 10.1016/j.ajpath.2020.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and cerebral amyloid angiopathy (CAA) are two distinct vascular angiopathies that share several similarities in clinical presentation and vascular pathology. Given the clinical and pathologic overlap, the molecular overlap between CADASIL and CAA was explored. CADASIL and CAA protein profiles from recently published proteomics-based and immuno-based studies were compared to investigate the potential for shared disease mechanisms. A comparison of affected proteins in each disease highlighted 19 proteins that are regulated in both CADASIL and CAA. Functional analysis of the shared proteins predicts significant interaction between them and suggests that most enriched proteins play roles in extracellular matrix structure and remodeling. Proposed models to explain the observed enrichment of extracellular matrix proteins include both increased protein secretion and decreased protein turnover by sequestration of chaperones and proteases or formation of stable protein complexes. Single-cell RNA sequencing of vascular cells in mice suggested that the vast majority of the genes accounting for the overlapped proteins between CADASIL and CAA are expressed by fibroblasts. Thus, our current understanding of the molecular profiles of CADASIL and CAA appears to support potential for common mechanisms underlying the two disorders.
Collapse
Affiliation(s)
- Kelly Z Young
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Gang Xu
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Simon G Keep
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Jimo Borjigin
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Michael M Wang
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan.
| |
Collapse
|
11
|
Howe MD, McCullough LD, Urayama A. The Role of Basement Membranes in Cerebral Amyloid Angiopathy. Front Physiol 2020; 11:601320. [PMID: 33329053 PMCID: PMC7732667 DOI: 10.3389/fphys.2020.601320] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Dementia is a neuropsychiatric syndrome characterized by cognitive decline in multiple domains, often leading to functional impairment in activities of daily living, disability, and death. The most common causes of age-related progressive dementia include Alzheimer's disease (AD) and vascular cognitive impairment (VCI), however, mixed disease pathologies commonly occur, as epitomized by a type of small vessel pathology called cerebral amyloid angiopathy (CAA). In CAA patients, the small vessels of the brain become hardened and vulnerable to rupture, leading to impaired neurovascular coupling, multiple microhemorrhage, microinfarction, neurological emergencies, and cognitive decline across multiple functional domains. While the pathogenesis of CAA is not well understood, it has long been thought to be initiated in thickened basement membrane (BM) segments, which contain abnormal protein deposits and amyloid-β (Aβ). Recent advances in our understanding of CAA pathogenesis link BM remodeling to functional impairment of perivascular transport pathways that are key to removing Aβ from the brain. Dysregulation of this process may drive CAA pathogenesis and provides an important link between vascular risk factors and disease phenotype. The present review summarizes how the structure and composition of the BM allows for perivascular transport pathways to operate in the healthy brain, and then outlines multiple mechanisms by which specific dementia risk factors may promote dysfunction of perivascular transport pathways and increase Aβ deposition during CAA pathogenesis. A better understanding of how BM remodeling alters perivascular transport could lead to novel diagnostic and therapeutic strategies for CAA patients.
Collapse
Affiliation(s)
| | | | - Akihiko Urayama
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
12
|
Identifying transglutaminase reaction products via mass spectrometry as exemplified by the MUC2 mucin - Pitfalls and traps. Anal Biochem 2020; 597:113668. [PMID: 32222540 PMCID: PMC7184670 DOI: 10.1016/j.ab.2020.113668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/15/2020] [Accepted: 02/27/2020] [Indexed: 12/26/2022]
Abstract
In order to demonstrate transglutaminase activity in biological samples immunological as well as glutamine- and amine-donor based assays are commonly used. However, the identification of the transglutaminase reaction product, i. e. the isopeptide cross-linked peptides/proteins or the deamidated protein/peptide are often neglected. This article describes a workflow for the detection of the products of transglutaminase-catalyzed reactions. In particular, possible pitfalls and traps that can arise during the mass spectrometry-based identification of isopeptide cross-links are addressed and characterised on actual samples.
Collapse
|
13
|
Wilhelmus MMM, Jongenelen CA, Bol JGJM, Drukarch B. Interaction between tissue transglutaminase and amyloid-beta: Protein-protein binding versus enzymatic crosslinking. Anal Biochem 2020; 592:113578. [PMID: 31923381 DOI: 10.1016/j.ab.2020.113578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/06/2020] [Indexed: 11/26/2022]
Abstract
Self-interaction, chaperone binding and posttranslational modification of amyloid-beta (Aβ) is essential in the initiation and propagation of Aβ aggregation. Aggregation results in insoluble Aβ deposits characteristic of Alzheimer's disease (AD) brain lesions, i.e. senile plaques and cerebral amyloid angiopathy. Tissue transglutaminase (tTG) is a calcium-dependent enzyme that catalyzes posttranslational modifications including the formation of covalent ε-(γ-glutamyl)lysine isopeptide bonds (molecular crosslinks), and colocalizes with Aβ deposits in AD. Two independent groups recently found that apart from the induction of Aβ oligomerization, the blood-derived transglutaminase member FXIIIa forms stable protein-protein complexes with Aβ independent of the transamidation reaction. Here, we investigated whether also tTG forms rigid protein complexes with Aβ in the absence of catalytic activation. We found that both Aβ1-40 and Aβ1-42 are substrates for tTG-catalyzed crosslinking. In addition, in the absence of calcium or the presence of a peptidergic inhibitor of tTG, stable tTG-Aβ1-40 complexes were found. Interestingly, the stable complexes between tTG and Aβ1-40, were only found at 'physiological' concentrations of Aβ1-40. Together, our data suggest that depending on the Aβ species at hand, and on the concentration of Aβ, rigid protein-complexes are formed between tTG and Aβ1-40 without the involvement of the crosslinking reaction.
Collapse
Affiliation(s)
- Micha M M Wilhelmus
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, the Netherlands.
| | - Cornelis A Jongenelen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, the Netherlands
| | - John G J M Bol
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, the Netherlands
| | - Benjamin Drukarch
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Bres EE, Faissner A. Low Density Receptor-Related Protein 1 Interactions With the Extracellular Matrix: More Than Meets the Eye. Front Cell Dev Biol 2019; 7:31. [PMID: 30931303 PMCID: PMC6428713 DOI: 10.3389/fcell.2019.00031] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a biological substrate composed of collagens, proteoglycans and glycoproteins that ensures proper cell migration and adhesion and keeps the cell architecture intact. The regulation of the ECM composition is a vital process strictly controlled by, among others, proteases, growth factors and adhesion receptors. As it appears, ECM remodeling is also essential for proper neuronal and glial development and the establishment of adequate synaptic signaling. Hence, disturbances in ECM functioning are often present in neurodegenerative diseases like Alzheimer’s disease. Moreover, mutations in ECM molecules are found in some forms of epilepsy and malfunctioning of ECM-related genes and pathways can be seen in, for example, cancer or ischemic injury. Low density lipoprotein receptor-related protein 1 (Lrp1) is a member of the low density lipoprotein receptor family. Lrp1 is involved not only in ligand uptake, receptor mediated endocytosis and lipoprotein transport—functions shared by low density lipoprotein receptor family members—but also regulates cell surface protease activity, controls cellular entry and binding of toxins and viruses, protects against atherosclerosis and acts on many cell signaling pathways. Given the plethora of functions, it is not surprising that Lrp1 also impacts the ECM and is involved in its remodeling. This review focuses on the role of Lrp1 and some of its major ligands on ECM function. Specifically, interactions with two Lrp1 ligands, integrins and tissue plasminogen activator are described in more detail.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
15
|
Abstract
Tissue transglutaminase (tTG), also referred to as type 2 transglutaminase or Gαh, can bind and hydrolyze GTP, as well as function as a protein crosslinking enzyme. tTG is widely expressed and can be detected both inside cells and in the extracellular space. In contrast to many enzymes, the active and inactive conformations of tTG are markedly different. The catalytically inactive form of tTG adopts a compact “closed-state” conformation, while the catalytically active form of the protein adopts an elongated “open-state” conformation. tTG has long been appreciated as an important player in numerous diseases, including celiac disease, neuronal degenerative diseases, and cancer, and its roles in these diseases often depend as much upon its conformation as its catalytic activity. While its ability to promote these diseases has been traditionally thought to be dependent on its protein crosslinking activity, more recent findings suggest that the conformational state tTG adopts is also important for mediating its effects. In particular, we and others have shown that the closed-state of tTG is important for promoting cell growth and survival, while maintaining tTG in the open-state is cytotoxic. In this review, we examine the two unique conformations of tTG and how they contribute to distinct biological processes. We will also describe how this information can be used to generate novel therapies to treat diseases, with a special focus on cancer.
Collapse
|
16
|
Ismail T, Vancha SR, Kanapathipillai M. L‐proline and betaine inhibit extracellular enzymes mediated abeta 1‐42 aggregation, oxidative stress, and toxicity. Pept Sci (Hoboken) 2018. [DOI: 10.1002/pep2.24093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Tania Ismail
- University of Michigan‐DearbornDepartment of Mechanical Engineering Dearborn Michigan
| | - Sushma Reddy Vancha
- University of Michigan‐DearbornDepartment of Mechanical Engineering Dearborn Michigan
| | | |
Collapse
|
17
|
Cystamine and cysteamine as inhibitors of transglutaminase activity in vivo. Biosci Rep 2018; 38:BSR20180691. [PMID: 30054429 PMCID: PMC6123069 DOI: 10.1042/bsr20180691] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/07/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cystamine is commonly used as a transglutaminase inhibitor. This disulphide undergoes reduction in vivo to the aminothiol compound, cysteamine. Thus, the mechanism by which cystamine inhibits transglutaminase activity in vivo could be due to either cystamine or cysteamine, which depends on the local redox environment. Cystamine inactivates transglutaminases by promoting the oxidation of two vicinal cysteine residues on the enzyme to an allosteric disulphide, whereas cysteamine acts as a competitive inhibitor for transamidation reactions catalyzed by this enzyme. The latter mechanism is likely to result in the formation of a unique biomarker, N-(γ-glutamyl)cysteamine that could serve to indicate how cyst(e)amine acts to inhibit transglutaminases inside cells and the body.
Collapse
|
18
|
Min B, Chung KC. New insight into transglutaminase 2 and link to neurodegenerative diseases. BMB Rep 2018; 51:5-13. [PMID: 29187283 PMCID: PMC5796628 DOI: 10.5483/bmbrep.2018.51.1.227] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Indexed: 12/13/2022] Open
Abstract
Formation of toxic protein aggregates is a common feature and mainly contributes to the pathogenesis of neurodegenerative diseases (NDDs), which include amyotrophic lateral sclerosis (ALS), Alzheimer’s, Parkinson’s, Huntington’s, and prion diseases. The transglutaminase 2 (TG2) gene encodes a multifunctional enzyme, displaying four types of activity, such as transamidation, GTPase, protein disulfide isomerase, and protein kinase activities. Many studies demonstrated that the calcium-dependent transamidation activity of TG2 affects the formation of insoluble and toxic amyloid aggregates that mainly consisted of NDD-related proteins. So far, many important and NDD-related substrates of TG2 have been identified, including amlyoid-β, tau, α-synuclein, mutant huntingtin, and ALS-linked trans-activation response (TAR) DNA-binding protein 43. Recently, the formation of toxic inclusions mediated by several TG2 substrates were efficiently inhibited by TG2 inhibitors. Therefore, the development of highly specific TG2 inhibitors would be an important tool in alleviating the progression of TG2-related brain disorders. In this review, the authors discuss recent advances in TG2 biochemistry, several mechanisms of molecular regulation and pleotropic signaling functions, and the presumed role of TG2 in the progression of many NDDs.
Collapse
Affiliation(s)
- Boram Min
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
19
|
Chrobok NL, Bol JGJM, Jongenelen CA, Brevé JJP, El Alaoui S, Wilhelmus MMM, Drukarch B, van Dam AM. Characterization of Transglutaminase 2 activity inhibitors in monocytes in vitro and their effect in a mouse model for multiple sclerosis. PLoS One 2018; 13:e0196433. [PMID: 29689097 PMCID: PMC5918173 DOI: 10.1371/journal.pone.0196433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023] Open
Abstract
The neurodegenerative disease multiple sclerosis (MS) is pathologically characterized by the massive influx of immune cells into the central nervous system. This contributes to demyelination and axonal damage which causes symptoms such as motor and cognitive dysfunctions. The migration of leukocytes from the blood vessel is orchestrated by a multitude of factors whose determination is essential in reducing cellular influx in MS patients and the experimental autoimmune encephalomyelitis (EAE) animal model. The here studied enzyme tissue Transglutaminase (TG2) is present intracellularly, on the cell surface and extracellularly. There it contributes to cellular adhesion and migration via its transamidation activity and possibly by facilitating cellular interaction with the extracellular matrix. Previous data from our group showed reduced motor symptoms and cellular infiltration after using a pharmacological TG2 transamidation activity inhibitor in a rat EAE model. However, it remained elusive if the cross-linking activity of the enzyme resulted in the observed effects. To follow-up, we now characterized two new small molecule TG2 activity inhibitors, BJJF078 and ERW1041E. Both compounds are potent inhibitor of recombinant human and mouse Transglutaminase enzyme activity, mainly TG2 and the close related enzyme TG1. In addition they did not affect the binding of TG2 to the extracellular matrix substrate fibronectin, a process via which TG2 promotes cellular adhesion and migration. We found, that ERW1041E but not BJJF078 resulted in reduced EAE disease motor-symptoms while neither caused apparent changes in pathology (cellular influx), Transglutaminase activity or expression of inflammation related markers in the spinal cord, compared to vehicle treated controls. Although we cannot exclude issues on bioavailability and in vivo efficacy of the used compounds, we hypothesize that extracellular TG1/TG2 activity is of greater importance than (intra-)cellular activity in mouse EAE pathology.
Collapse
Affiliation(s)
- Navina L. Chrobok
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - John G. J. M. Bol
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Cornelis A. Jongenelen
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - John J. P. Brevé
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Micha M. M. Wilhelmus
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Grand Moursel L, van Roon-Mom WMC, Kiełbasa SM, Mei H, Buermans HPJ, van der Graaf LM, Hettne KM, de Meijer EJ, van Duinen SG, Laros JFJ, van Buchem MA, 't Hoen PAC, van der Maarel SM, van der Weerd L. Brain Transcriptomic Analysis of Hereditary Cerebral Hemorrhage With Amyloidosis-Dutch Type. Front Aging Neurosci 2018; 10:102. [PMID: 29706885 PMCID: PMC5908973 DOI: 10.3389/fnagi.2018.00102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/26/2018] [Indexed: 11/23/2022] Open
Abstract
Hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) caused by a point mutation resulting in an amino acid change (NP_000475.1:p.Glu693Gln) in the amyloid precursor protein (APP). Post-mortem frontal and occipital cortical brain tissue from nine patients and nine age-related controls was used for RNA sequencing to identify biological pathways affected in HCHWA-D. Although previous studies indicated that pathology is more severe in the occipital lobe in HCHWA-D compared to the frontal lobe, the current study showed similar changes in gene expression in frontal and occipital cortex and the two brain regions were pooled for further analysis. Significantly altered pathways were analyzed using gene set enrichment analysis (GSEA) on 2036 significantly differentially expressed genes. Main pathways over-represented by down-regulated genes were related to cellular aerobic respiration (including ATP synthesis and carbon metabolism) indicating a mitochondrial dysfunction. Principal up-regulated pathways were extracellular matrix (ECM)–receptor interaction and ECM proteoglycans in relation with an increase in the transforming growth factor beta (TGFβ) signaling pathway. Comparison with the publicly available dataset from pre-symptomatic APP-E693Q transgenic mice identified overlap for the ECM–receptor interaction pathway, indicating that ECM modification is an early disease specific pathomechanism.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Szymon M Kiełbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, Netherlands
| | - Hailiang Mei
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, Netherlands
| | - Henk P J Buermans
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Kristina M Hettne
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Emile J de Meijer
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Jeroen F J Laros
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
21
|
Pita-Juárez Y, Altschuler G, Kariotis S, Wei W, Koler K, Green C, Tanzi RE, Hide W. The Pathway Coexpression Network: Revealing pathway relationships. PLoS Comput Biol 2018; 14:e1006042. [PMID: 29554099 PMCID: PMC5875878 DOI: 10.1371/journal.pcbi.1006042] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 03/29/2018] [Accepted: 02/19/2018] [Indexed: 02/02/2023] Open
Abstract
A goal of genomics is to understand the relationships between biological processes. Pathways contribute to functional interplay within biological processes through complex but poorly understood interactions. However, limited functional references for global pathway relationships exist. Pathways from databases such as KEGG and Reactome provide discrete annotations of biological processes. Their relationships are currently either inferred from gene set enrichment within specific experiments, or by simple overlap, linking pathway annotations that have genes in common. Here, we provide a unifying interpretation of functional interaction between pathways by systematically quantifying coexpression between 1,330 canonical pathways from the Molecular Signatures Database (MSigDB) to establish the Pathway Coexpression Network (PCxN). We estimated the correlation between canonical pathways valid in a broad context using a curated collection of 3,207 microarrays from 72 normal human tissues. PCxN accounts for shared genes between annotations to estimate significant correlations between pathways with related functions rather than with similar annotations. We demonstrate that PCxN provides novel insight into mechanisms of complex diseases using an Alzheimer’s Disease (AD) case study. PCxN retrieved pathways significantly correlated with an expert curated AD gene list. These pathways have known associations with AD and were significantly enriched for genes independently associated with AD. As a further step, we show how PCxN complements the results of gene set enrichment methods by revealing relationships between enriched pathways, and by identifying additional highly correlated pathways. PCxN revealed that correlated pathways from an AD expression profiling study include functional clusters involved in cell adhesion and oxidative stress. PCxN provides expanded connections to pathways from the extracellular matrix. PCxN provides a powerful new framework for interrogation of global pathway relationships. Comprehensive exploration of PCxN can be performed at http://pcxn.org/. Genes do not function alone, but interact within pathways to carry out specific biological processes. Pathways, in turn, interact at a higher level to affect major cellular activities such as motility, growth and development. We present a pathway coexpression network (PCxN) that systematically maps and quantifies these high-level interactions and establishes a unifying reference for pathway relationships. The method uses 3,207 human microarrays from 72 normal human tissues and 1,330 of the most well established pathway annotations to describe global relationships between pathways. PCxN accounts for shared genes to estimate correlations between pathways with related functions rather than with redundant pathway definitions. PCxN can be used to discover and explore pathways correlated with a pathway of interest. We applied PCxN to identify key processes related to Alzheimer’s disease (AD), interpreting a mixed genetic association and experimental derived set of disease genes in the context of gene co-expression. We expand the known relationships between pathways identified by gene set enrichment analysis in brain tissues affected with AD. PCxN provides a high-level overview of pathway relationships. PCxN is available as a webtool at http://pcxn.org/, and as a Bioconductor package at http://bioconductor.org/packages/pcxn/.
Collapse
Affiliation(s)
- Yered Pita-Juárez
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, United States of America
| | - Gabriel Altschuler
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Sokratis Kariotis
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Wenbin Wei
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Katjuša Koler
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Claire Green
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Winston Hide
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, United States of America
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
- Harvard Stem Cell Institute, Cambridge, Massachusetts, United States of America
- National Institute Health Research, Sheffield Biomedical Research Centre, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
Identification of post-translational modifications of Aβ peptide in platelet membranes from patients with cerebral amyloid angiopathy. J Neurol Sci 2017; 383:11-17. [DOI: 10.1016/j.jns.2017.08.3269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023]
|
23
|
Grand Moursel L, Munting LP, van der Graaf LM, van Duinen SG, Goumans MJTH, Ueberham U, Natté R, van Buchem MA, van Roon-Mom WMC, van der Weerd L. TGFβ pathway deregulation and abnormal phospho-SMAD2/3 staining in hereditary cerebral hemorrhage with amyloidosis-Dutch type. Brain Pathol 2017; 28:495-506. [PMID: 28557134 PMCID: PMC8028662 DOI: 10.1111/bpa.12533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/19/2017] [Indexed: 12/20/2022] Open
Abstract
Hereditary cerebral hemorrhage with amyloidosis‐Dutch type (HCHWA‐D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) pathology, caused by the E22Q mutation in the amyloid β (Aβ) peptide. Transforming growth factor β1 (TGFβ1) is a key player in vascular fibrosis and in the formation of angiopathic vessels in transgenic mice. Therefore, we investigated whether the TGFβ pathway is involved in HCHWA‐D pathogenesis in human postmortem brain tissue from frontal and occipital lobes. Components of the TGFβ pathway were analyzed with quantitative RT‐PCR. TGFβ1 and TGFβ Receptor 2 (TGFBR2) gene expression levels were significantly increased in HCHWA‐D in comparison to the controls, in both frontal and occipital lobes. TGFβ‐induced pro‐fibrotic target genes were also upregulated. We further assessed pathway activation by detecting phospho‐SMAD2/3 (pSMAD2/3), a direct TGFβ down‐stream signaling mediator, using immunohistochemistry. We found abnormal pSMAD2/3 granular deposits specifically on HCHWA‐D angiopathic frontal and occipital vessels. We graded pSMAD2/3 accumulation in angiopathic vessels and found a positive correlation with the CAA load independent of the brain area. We also observed pSMAD2/3 granules in a halo surrounding occipital vessels, which was specific for HCHWA‐D. The result of this study indicates an upregulation of TGFβ1 in HCHWA‐D, as was found previously in AD with CAA pathology. We discuss the possible origins and implications of the TGFβ pathway deregulation in the microvasculature in HCHWA‐D. These findings identify the TGFβ pathway as a potential biomarker of disease progression and a possible target of therapeutic intervention in HCHWA‐D.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Leon P Munting
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Uwe Ueberham
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Remco Natté
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
24
|
Kawabe K, Takano K, Moriyama M, Nakamura Y. Transglutaminases Derived from Astrocytes Accelerate Amyloid β Aggregation. Neurochem Res 2017; 42:2384-2391. [DOI: 10.1007/s11064-017-2258-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
|
25
|
Rudinskiy N, Fuerer C, Demurtas D, Zamorano S, De Piano C, Herrmann AG, Spires-Jones TL, Oeckl P, Otto M, Frosch MP, Moniatte M, Hyman BT, Schmid AW. Amyloid‐beta oligomerization is associated with the generation of a typical peptide fragment fingerprint. Alzheimers Dement 2016; 12:996-1013. [DOI: 10.1016/j.jalz.2016.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 02/29/2016] [Accepted: 03/19/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Nikita Rudinskiy
- Department of Neurology, Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital Harvard Medical School Charlestown MA USA
| | - Christophe Fuerer
- School of Life Sciences, Proteomics Core Facility Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Davide Demurtas
- School of Basic Sciences, Interdisciplinary Centre for Electron Microscopy (CIME) Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Sebastian Zamorano
- School of Life Sciences, Proteomics Core Facility Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Cyntia De Piano
- School of Life Sciences, Proteomics Core Facility Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Abigail G. Herrmann
- Center for Cognitive and Neural Systems The University of Edinburgh Edinburgh Scotland
- Centre for Dementia Prevention The University of Edinburgh Edinburgh Scotland
- Euan MacDonald Centre for Motorneurone Disease The University of Edinburgh Edinburgh Scotland
| | - Tara L. Spires-Jones
- Center for Cognitive and Neural Systems The University of Edinburgh Edinburgh Scotland
- Centre for Dementia Prevention The University of Edinburgh Edinburgh Scotland
- Euan MacDonald Centre for Motorneurone Disease The University of Edinburgh Edinburgh Scotland
| | - Patrick Oeckl
- Department of Neurology Ulm University Hospital Ulm Germany
| | - Markus Otto
- Department of Neurology Ulm University Hospital Ulm Germany
| | - Matthew P. Frosch
- Massachusetts General Hospital and Harvard Medical School Massachusetts General Institute for Neurodegenerative Disease Charlestown MA USA
| | - Marc Moniatte
- School of Life Sciences, Proteomics Core Facility Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Bradley T. Hyman
- Department of Neurology, Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital Harvard Medical School Charlestown MA USA
| | - Adrien W. Schmid
- School of Life Sciences, Proteomics Core Facility Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| |
Collapse
|
26
|
Wilhelmus MMM, de Jager M, Smit AB, van der Loo RJ, Drukarch B. Catalytically active tissue transglutaminase colocalises with Aβ pathology in Alzheimer's disease mouse models. Sci Rep 2016; 6:20569. [PMID: 26837469 PMCID: PMC4738336 DOI: 10.1038/srep20569] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is characterised by amyloid-beta (Aβ) protein deposition in the brain. Posttranslational modifications in Aβ play an important role in Aβ deposition. Tissue transglutaminase (tTG) is an enzyme involved in posttranslational cross-linking of proteins. tTG levels and activity are increased in AD brains, and tTG is associated with Aβ deposits and lesion-associated astrocytes in AD cases. Furthermore, Aβ is a substrate of tTG-catalysed cross-linking. To study the role of tTG in Aβ pathology, we compared tTG distribution and activity in both the APPSWE/PS1ΔE9 and APP23 mice models with human AD. Using immunohistochemistry, we found association of both tTG and in situ active tTG with Aβ plaques and vascular Aβ, in early and late stages of Aβ deposition. In addition, tTG staining colocalised with Aβ-associated reactive astrocytes. Thus, alike human AD cases, tTG was associated with Aβ depositions in these AD models. Although, distribution pattern and spatial overlay of both tTG and its activity with Aβ pathology was substantially different from human AD cases, our findings provide evidence for an early role of tTG in Aβ pathology. Yet, species differences should be taken into account when using these models to study the role of tTG in Aβ pathology.
Collapse
Affiliation(s)
- Micha M M Wilhelmus
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU medical center, Amsterdam, The Netherlands
| | - Mieke de Jager
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU medical center, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU medical center, Amsterdam, The Netherlands
| |
Collapse
|
27
|
van der Wildt B, Wilhelmus MMM, Bijkerk J, Haveman LYF, Kooijman EJM, Schuit RC, Bol JGJM, Jongenelen CAM, Lammertsma AA, Drukarch B, Windhorst AD. Development of carbon-11 labeled acryl amides for selective PET imaging of active tissue transglutaminase. Nucl Med Biol 2016; 43:232-42. [PMID: 27067043 DOI: 10.1016/j.nucmedbio.2016.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/04/2016] [Accepted: 01/16/2016] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Tissue transglutaminase (TG2) is a ubiquitously expressed enzyme capable of forming metabolically and mechanically stable crosslinks between the γ-carboxamide of a glutamine acyl-acceptor substrate and the ε-amino functionality of a lysine acyl-donor substrate resulting in protein oligomers. High TG2 crosslinking activity has been implicated in the pathogenesis of various diseases including celiac disease, cancer and fibrotic and neurodegenerative diseases. Development of a PET tracer specific for active TG2 provides a novel tool to further investigate TG2 biology in vivo in disease states. Recently, potent irreversible active site TG2 inhibitors carrying an acrylamide warhead were synthesized and pharmacologically characterized. METHODS Three of these inhibitors, compound 1, 2 and 3, were successfully radiolabeled with carbon-11 on the acrylamide carbonyl position using a palladium mediated [(11)C]CO aminocarbonylation reaction. Ex vivo biodistribution and plasma stability were evaluated in healthy Wistar rats. Autoradiography was performed on MDA-MB-231 tumor sections. RESULTS [(11)C]1, -2 and -3 were obtained in decay corrected radiochemical yields of 38-55%. Biodistribution showed low uptake in peripheral tissues, with the exception of liver and kidney. Low brain uptake of <0.05% ID/g was observed. Blood plasma analysis demonstrated that [(11)C]1 and [(11)C]2 were rapidly metabolized, whereas [(11)C]3 was metabolized at a more moderate rate (63.2 ± 6.8 and 28.7 ± 10.8% intact tracer after 15 and 45 min, respectively). Autoradiography with [(11)C]3 on MDA-MB-231 tumor sections showed selective and specific binding of the radiotracer to the active state of TG2. CONCLUSIONS Taken together, these results identify [(11)C]3 as the most promising of the three compounds tested for development as PET radiotracer for the in vivo investigation of TG2 activity.
Collapse
Affiliation(s)
- Berend van der Wildt
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; Departments of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands.
| | - Micha M M Wilhelmus
- Departments of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jonne Bijkerk
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Lizeth Y F Haveman
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Esther J M Kooijman
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert C Schuit
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - John G J M Bol
- Departments of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Cornelis A M Jongenelen
- Departments of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Drukarch
- Departments of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Departments of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
28
|
de Jager M, Drukarch B, Hofstee M, Brevé J, Jongenelen CAM, Bol JGJM, Wilhelmus MMM. Tissue transglutaminase-catalysed cross-linking induces Apolipoprotein E multimers inhibiting Apolipoprotein E's protective effects towards amyloid-beta-induced toxicity. J Neurochem 2015; 134:1116-28. [DOI: 10.1111/jnc.13203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/12/2015] [Accepted: 06/08/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Mieke de Jager
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| | - Marloes Hofstee
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| | - John Brevé
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| | - Cornelis A. M. Jongenelen
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| | - John G. J. M. Bol
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| | - Micha M. M. Wilhelmus
- Department of Anatomy and Neurosciences; Neuroscience Campus Amsterdam; VU University Medical Center; Amsterdam The Netherlands
| |
Collapse
|
29
|
de Jager M, Boot MV, Bol JGJM, Brevé JJP, Jongenelen CAM, Drukarch B, Wilhelmus MMM. The blood clotting Factor XIIIa forms unique complexes with amyloid-beta (Aβ) and colocalizes with deposited Aβ in cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 2015; 42:255-72. [PMID: 25871449 DOI: 10.1111/nan.12244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/09/2015] [Indexed: 12/11/2022]
Abstract
AIMS Cerebral amyloid angiopathy (CAA) is a key pathological hallmark of Alzheimer's disease (AD) characterized by accumulation of amyloid-beta (Aβ) protein in blood vessel walls. CAA impairs vessel functioning, affects blood brain barrier integrity and accelerates cognitive decline of AD patients. Unfortunately, mechanisms underlying Aβ deposition in the vessel wall remain largely unknown. Factor XIIIa (FXIIIa) is a blood-derived transglutaminase crucial in blood coagulation by cross-linking fibrin molecules. Evidence is mounting that blood-derived factors are present in CAA and may play a role in protein deposition in the vessel wall. We therefore investigated whether FXIIIa is present in CAA and if FXIIIa cross-link activity affects Aβ aggregation. METHODS Using immunohistochemistry, we investigated the distribution of FXIIIa, its activator thrombin and in situ FXIIIa activity in CAA in post-mortem AD tissue. We used surface plasmon resonance and Western blot analysis to study binding of FXIIIa to Aβ and the formation of FXIIIa-Aβ complexes, respectively. In addition, we studied cytotoxicity of FXIIIa-Aβ complexes to cerebrovascular cells. RESULTS FXIIIa, thrombin and in situ FXIIIa activity colocalize with the Aβ deposition in CAA. Furthermore, FXIIIa binds to Aβ with a higher binding affinity for Aβ1-42 compared with Aβ1-40 . Moreover, highly stable FXIIIa-Aβ complexes are formed independently of FXIIIa cross-linking activity that protected cerebrovascular cells from Aβ-induced toxicity in vitro. CONCLUSIONS Our data showed that FXIIIa colocalizes with Aβ in CAA and that FXIIIa forms unique protein complexes with Aβ that might play an important role in Aβ deposition and persistence in the vessel wall.
Collapse
Affiliation(s)
- M de Jager
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - M V Boot
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - J G J M Bol
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - J J P Brevé
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - C A M Jongenelen
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - B Drukarch
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - M M M Wilhelmus
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Espitia Pinzon N, Stroo E, ‘t Hart BA, Bol JGJM, Drukarch B, Bauer J, van Dam AM. Tissue transglutaminase in marmoset experimental multiple sclerosis: discrepancy between white and grey matter. PLoS One 2014; 9:e100574. [PMID: 24959868 PMCID: PMC4069090 DOI: 10.1371/journal.pone.0100574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/26/2014] [Indexed: 02/06/2023] Open
Abstract
Infiltration of leukocytes is a major pathological event in white matter lesion formation in the brain of multiple sclerosis (MS) patients. In grey matter lesions, less infiltration of these cells occur, but microglial activation is present. Thus far, the interaction of β-integrins with extracellular matrix proteins, e.g. fibronectin, is considered to be of importance for the influx of immune cells. Recent in vitro studies indicate a possible role for the enzyme tissue Transglutaminase (TG2) in mediating cell adhesion and migration. In the present study we questioned whether TG2 is present in white and grey matter lesions observed in the marmoset model for MS. To this end, immunohistochemical studies were performed. We observed that TG2, expressed by infiltrating monocytes in white matter lesions co-expressed β1-integrin and is located in close apposition to deposited fibronectin. These data suggest an important role for TG2 in the adhesion and migration of infiltrating monocytes during white matter lesion formation. Moreover, in grey matter lesions, TG2 is mainly present in microglial cells together with some β1-integrin, whereas fibronectin is absent in these lesions. These data imply an alternative role for microglial-derived TG2 in grey matter lesions, e.g. cell proliferation. Further research should clarify the functional role of TG2 in monocytes or microglial cells in MS lesion formation.
Collapse
Affiliation(s)
- Nathaly Espitia Pinzon
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Esther Stroo
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Bert A. ‘t Hart
- Biomedical Primate Research Center, Department of Immunobiology, Rijswijk, The Netherlands
- University Groningen, University Medical Center, Department of Neuroscience, Groningen, The Netherlands
| | - John G. J. M. Bol
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Benjamin Drukarch
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Jan Bauer
- Center for Brain Research, Department of Neuroimmunology, Vienna, Austria
| | - Anne-Marie van Dam
- VU University Medical Center, Neuroscience Campus Amsterdam, Department of Anatomy and Neurosciences, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
31
|
Lathe R, Sapronova A, Kotelevtsev Y. Atherosclerosis and Alzheimer--diseases with a common cause? Inflammation, oxysterols, vasculature. BMC Geriatr 2014; 14:36. [PMID: 24656052 PMCID: PMC3994432 DOI: 10.1186/1471-2318-14-36] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/26/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Aging is accompanied by increasing vulnerability to pathologies such as atherosclerosis (ATH) and Alzheimer disease (AD). Are these different pathologies, or different presentations with a similar underlying pathoetiology? DISCUSSION Both ATH and AD involve inflammation, macrophage infiltration, and occlusion of the vasculature. Allelic variants in common genes including APOE predispose to both diseases. In both there is strong evidence of disease association with viral and bacterial pathogens including herpes simplex and Chlamydophila. Furthermore, ablation of components of the immune system (or of bone marrow-derived macrophages alone) in animal models restricts disease development in both cases, arguing that both are accentuated by inflammatory/immune pathways. We discuss that amyloid β, a distinguishing feature of AD, also plays a key role in ATH. Several drugs, at least in mouse models, are effective in preventing the development of both ATH and AD. Given similar age-dependence, genetic underpinnings, involvement of the vasculature, association with infection, Aβ involvement, the central role of macrophages, and drug overlap, we conclude that the two conditions reflect different manifestations of a common pathoetiology. MECHANISM Infection and inflammation selectively induce the expression of cholesterol 25-hydroxylase (CH25H). Acutely, the production of 'immunosterol' 25-hydroxycholesterol (25OHC) defends against enveloped viruses. We present evidence that chronic macrophage CH25H upregulation leads to catalyzed esterification of sterols via 25OHC-driven allosteric activation of ACAT (acyl-CoA cholesterol acyltransferase/SOAT), intracellular accumulation of cholesteryl esters and lipid droplets, vascular occlusion, and overt disease. SUMMARY We postulate that AD and ATH are both caused by chronic immunologic challenge that induces CH25H expression and protection against particular infectious agents, but at the expense of longer-term pathology.
Collapse
Affiliation(s)
- Richard Lathe
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Pieta Research, PO Box 27069, Edinburgh EH10 5YW, UK
| | - Alexandra Sapronova
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Optical Research Group, Laboratory of Evolutionary Biophysics of Development, Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Yuri Kotelevtsev
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Biomedical Centre for Research Education and Innovation (CREI), Skolkovo Institute of Science and Technology, Skolkovo 143025, Russia
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Little France, Edinburgh EH16 4TJ, UK
| |
Collapse
|
32
|
Characterization of SLCO5A1/OATP5A1, a solute carrier transport protein with non-classical function. PLoS One 2013; 8:e83257. [PMID: 24376674 PMCID: PMC3869781 DOI: 10.1371/journal.pone.0083257] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 11/01/2013] [Indexed: 12/14/2022] Open
Abstract
Organic anion transporting polypeptides (OATP/SLCO) have been identified to mediate the uptake of a broad range of mainly amphipathic molecules. Human OATP5A1 was found to be expressed in the epithelium of many cancerous and non-cancerous tissues throughout the body but protein characterization and functional analysis have not yet been performed. This study focused on the biochemical characterization of OATP5A1 using Xenopus laevis oocytes and Flp-In T-REx-HeLa cells providing evidence regarding a possible OATP5A1 function. SLCO5A1 is highly expressed in mature dendritic cells compared to immature dendritic cells (∼6.5-fold) and SLCO5A1 expression correlates with the differentiation status of primary blood cells. A core- and complex- N-glycosylated polypeptide monomer of ∼105 kDa and ∼130 kDa could be localized in intracellular membranes and on the plasma membrane, respectively. Inducible expression of SLCO5A1 in HeLa cells led to an inhibitory effect of ∼20% after 96 h on cell proliferation. Gene expression profiling with these cells identified immunologically relevant genes (e.g. CCL20) and genes implicated in developmental processes (e.g. TGM2). A single nucleotide polymorphism leading to the exchange of amino acid 33 (L→F) revealed no differences regarding protein expression and function. In conclusion, we provide evidence that OATP5A1 might be a non-classical OATP family member which is involved in biological processes that require the reorganization of the cell shape, such as differentiation and migration.
Collapse
|
33
|
Wilhelmus MMM, Drukarch B. Tissue transglutaminase is a biochemical marker for Alzheimer's disease. Neurobiol Aging 2013; 35:e3-4. [PMID: 24080177 DOI: 10.1016/j.neurobiolaging.2013.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/15/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
Affiliation(s)
- Micha M M Wilhelmus
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|