1
|
Young M, McLeod DSA, Richard K. Nicotine increases hepatocyte transthyretin turnover: A possible mechanism for the protective effect of smoking on preeclampsia? Mol Cell Endocrinol 2025; 597:112446. [PMID: 39725350 DOI: 10.1016/j.mce.2024.112446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Transthyretin is a thyroid hormone binding protein with a major role in the distribution of thyroid hormones to peripheral tissues. In preeclampsia, the failing placenta releases soluble endoglin into the maternal circulation causing systemic vascular dysfunction. Our group has previously shown that transthyretin binds to soluble endoglin and is taken up as a complex into hepatocytes. The risk of developing preeclampsia is greatly reduced by smoking cigarettes. The addictive component of cigarette smoke, nicotine, increases transthyretin expression in rodent brain and also stabilises binding between thyroxine and transthyretin. The aim of this study was to determine the effects of nicotine on transthyretin expression, secretion and uptake by hepatocytes and if nicotine altered the uptake of transthyretin bound soluble endoglin. Nicotine treatment increased transthyretin mRNA and protein levels in cultured hepatocytes. Using live cell imaging, Alexa-transthyretin uptake was significantly increased in the presence of nicotine. Alexa-soluble endoglin uptake was also significantly increased by exposure to nicotine. The transthyretin-Alexa-soluble endoglin complex was taken up via the low-density lipoprotein receptor related protein-1 (LRP1). LRP1 protein levels were unaffected in nicotine treated hepatocytes. Nicotine exposure increases hepatocyte synthesis, secretion and uptake of transthyretin as well as cell uptake of soluble endoglin. Nicotine may protect against preeclampsia by increasing serum TTR which can bind soluble endoglin and remove it from the circulation. Further research is required to better understand the role of transthyretin and nicotine in mitigating preeclampsia.
Collapse
Affiliation(s)
- Melanie Young
- Conjoint Endocrine Laboratory, Chemical Pathology, Pathology Queensland, Queensland Health, Herston, Qld 4029, Australia
| | - Donald S A McLeod
- Department of Endocrinology and Diabetes, Royal Brisbane and Women's Hospital, Herston, Qld 4029, Australia
| | - Kerry Richard
- Conjoint Endocrine Laboratory, Chemical Pathology, Pathology Queensland, Queensland Health, Herston, Qld 4029, Australia; School of Medicine, University of Queensland, Herston, Qld 4029, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, Qld 4000, Australia.
| |
Collapse
|
2
|
Lamisa AB, Ahammad I, Bhattacharjee A, Hossain MU, Ishtiaque A, Chowdhury ZM, Das KC, Salimullah M, Keya CA. A meta-analysis of bulk RNA-seq datasets identifies potential biomarkers and repurposable therapeutics against Alzheimer's disease. Sci Rep 2024; 14:24717. [PMID: 39433822 PMCID: PMC11494203 DOI: 10.1038/s41598-024-75431-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Alzheimer's disease (AD) poses a major challenge due to its impact on the elderly population and the lack of effective early diagnosis and treatment options. In an effort to address this issue, a study focused on identifying potential biomarkers and therapeutic agents for AD was carried out. Using RNA-Seq data from AD patients and healthy individuals, 12 differentially expressed genes (DEGs) were identified, with 9 expressing upregulation (ISG15, HRNR, MTATP8P1, MTCO3P12, DTHD1, DCX, ST8SIA2, NNAT, and PCDH11Y) and 3 expressing downregulation (LTF, XIST, and TTR). Among them, TTR exhibited the lowest gene expression profile. Interestingly, functional analysis tied TTR to amyloid fiber formation and neutrophil degranulation through enrichment analysis. These findings suggested the potential of TTR as a diagnostic biomarker for AD. Additionally, druggability analysis revealed that the FDA-approved drug Levothyroxine might be effective against the Transthyretin protein encoded by the TTR gene. Molecular docking and dynamics simulation studies of Levothyroxine and Transthyretin suggested that this drug could be repurposed to treat AD. However, additional studies using in vitro and in vivo models are necessary before these findings can be applied in clinical applications.
Collapse
Affiliation(s)
- Anika Bushra Lamisa
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, 1229, Bangladesh
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Ahmed Ishtiaque
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Savar, Dhaka, 1349, Ashulia, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, 1229, Bangladesh.
| |
Collapse
|
3
|
Tsoy A, Umbayev B, Kassenova A, Kaupbayeva B, Askarova S. Pathology of Amyloid-β (Aβ) Peptide Peripheral Clearance in Alzheimer's Disease. Int J Mol Sci 2024; 25:10964. [PMID: 39456746 PMCID: PMC11507512 DOI: 10.3390/ijms252010964] [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: 08/30/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Although Alzheimer's disease (AD) is traditionally viewed as a central nervous system disorder driven by the cerebral accumulation of toxic beta-amyloid (Aβ) peptide, new interpretations of the amyloid cascade hypothesis have led to the recognition of the dynamic equilibrium in which Aβ resides and the importance of peripheral Aβ production and degradation in maintaining healthy Aβ levels. Our review sheds light on the critical role of peripheral organs, particularly the liver, in the metabolism and clearance of circulating Aβ. We explore the mechanisms of Aβ transport across the blood-brain barrier (BBB) via transport proteins such as LRP1 and P-glycoprotein. We also examine how peripheral clearance mechanisms, including enzymatic degradation and phagocytic activity, impact Aβ homeostasis. Our review also discusses potential therapeutic strategies targeting peripheral Aβ clearance pathways. By enhancing these pathways, we propose a novel approach to reducing cerebral Aβ burden, potentially slowing AD progression.
Collapse
Affiliation(s)
- Andrey Tsoy
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.T.); (B.U.); (A.K.); (B.K.)
| | - Bauyrzhan Umbayev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.T.); (B.U.); (A.K.); (B.K.)
| | - Aliya Kassenova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.T.); (B.U.); (A.K.); (B.K.)
- Faculty of Natural Sciences, Eurasian National University, Astana 010000, Kazakhstan
| | - Bibifatima Kaupbayeva
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.T.); (B.U.); (A.K.); (B.K.)
| | - Sholpan Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.T.); (B.U.); (A.K.); (B.K.)
| |
Collapse
|
4
|
Sadanandan J, Sathyanesan M, Newton SS. Aging alters the expression of trophic factors and tight junction proteins in the mouse choroid plexus. Fluids Barriers CNS 2024; 21:77. [PMID: 39334352 PMCID: PMC11438291 DOI: 10.1186/s12987-024-00574-0] [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: 03/18/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The choroid plexus (CP) is an understudied tissue in the central nervous system and is primarily implicated in cerebrospinal fluid (CSF) production. CP also produces numerous neurotrophic factors (NTF) which circulate to different brain regions. Regulation of NTFs in the CP during natural aging is largely unknown. Here, we investigated the age and gender-specific transcription of NTFs along with the changes in the tight junctional proteins (TJPs) and the water channel protein Aquaporin (AQP1). METHODS Male and female mice were used for our study. Age-related transcriptional changes were analyzed using quantitative PCR at three different time points: mature adult, middle-aged, and aged. Transcriptional changes during aging were further confirmed with digital droplet PCR. Additionally, we used immunohistochemical analysis (IHC) for the evaluation of in vivo protein expression. We further investigated the cellular phenotype of these NTFS, TJP, and water channel proteins in the mouse CP by co-labeling them with the classical vascular marker, Isolectin B4, and epithelial cell marker, Plectin. RESULTS Aging significantly altered NTF gene expression in the CP. Brain-derived neurotrophic factor (BDNF), Midkine (MDK), VGF, Insulin-like growth factor (IGF1), IGF2, Klotho (KL), Erythropoietin (EPO), and its receptor (EPOR) were reduced in the aged CP of males and females. Vascular endothelial growth factor (VEGF) transcription was gender-specific; in males, gene expression was unchanged in the aged CP, while females showed an age-dependent reduction. Age-dependent changes in VEGF localization were evident, from vasculature to epithelial cells. IGF2 and klotho localized in the basolateral membrane of the CP and showed an age-dependent reduction in epithelial cells. Water channel protein AQP1 localized in the tip of epithelial cells and showed an age-related reduction in mRNA and protein levels. TJP's JAM, CLAUDIN1, CLAUDIN2 and CLAUDIN5 were reduced in aged mice. CONCLUSIONS Our study highlights transcriptional level changes in the CP during aging. The age-related transcriptional changes exhibit similarities as well as gene-specific differences in the CP of males and females. Altered transcription of the water channel protein AQP1 and TJPs could be involved in reduced CSF production during aging. Importantly, reduction in the neurotrophic factors and longevity factor Klotho can play a role in regulating brain aging.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
5
|
Corino C, Aimo A, Luigetti M, Ciccone L, Ferrari Chen YF, Panichella G, Musetti V, Castiglione V, Vergaro G, Emdin M, Franzini M. Tetrameric Transthyretin as a Protective Factor Against Alzheimer's Disease. Mol Neurobiol 2024:10.1007/s12035-024-04442-8. [PMID: 39192044 DOI: 10.1007/s12035-024-04442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
Transthyretin (TTR) is a tetrameric protein traditionally recognized for its role in transporting thyroxine and retinol. Recent research has highlighted the potential neuroprotective functions of TTR in the setting of Alzheimer's disease (AD), which is the most common form of dementia and is caused by the deposition of amyloid beta (Aβ) and the resulting cytotoxic effects. This paper explores the mechanisms of TTR protective action, including its interaction with Aβ to prevent fibril formation and promote Aβ clearance from the brain. It also synthesizes experimental evidence suggesting that enhanced TTR stability may mitigate neurodegeneration and cognitive decline in AD. Potential therapeutic strategies such as small molecule stabilizers of TTR are discussed, highlighting their role in enhancing TTR binding to Aβ and facilitating its clearance. By consolidating current knowledge and proposing directions for future research, this review aims to underscore the significance of TTR as a neuroprotective factor in AD and the potential implications for future research.
Collapse
Affiliation(s)
- Camilla Corino
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Alberto Aimo
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Marco Luigetti
- Fondazione Policlinico Agostino Gemelli IRCCS, UOC Neurologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lidia Ciccone
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Yu Fu Ferrari Chen
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Veronica Musetti
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Vincenzo Castiglione
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Maria Franzini
- Department of Translational Research On New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
6
|
Han SW, Lee SH, Kim JH, Lee JJ, Park YH, Kim S, Nho K, Sohn JH. Association of liver function markers and apolipoprotein E ε4 with pathogenesis and cognitive decline in Alzheimer's disease. Front Aging Neurosci 2024; 16:1411466. [PMID: 39114318 PMCID: PMC11303325 DOI: 10.3389/fnagi.2024.1411466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Background Alzheimer's disease (AD) is a complex neurodegenerative disorder influenced by various factors, including liver function, which may impact the clearance of amyloid-β (Aβ) in the brain. This study aimed to explore how the apolipoprotein E (APOE) ε4 allele affects the relationship of liver function markers with AD pathology and cognition. Methods We analyzed data from two independent cohorts, including 732 participants from the Hallym University Medical Center and 483 from the Alzheimer's Disease Neuroimaging Initiative, each group consisting of individuals with and without the APOE ε4 allele. Cross-sectional analyses evaluated the associations of liver enzymes (aspartate aminotransferase [AST], alanine aminotransferase [ALT], alkaline phosphatase, total bilirubin, and albumin) with AD diagnosis, amyloid positron emission tomography (PET) burden, and cerebrospinal fluid biomarkers for AD (Aβ42, total tau, and phosphorylated tau181) at baseline. Longitudinally, we investigated the associations between these liver enzymes and changes in cognitive performance over the course of a year. Logistic and linear regression models were used to analyze these associations and mediation analyses were conducted to assess whether age and amyloid PET burden mediated these associations. Results Only in the APOE ε4 carrier group, a high AST to ALT ratio and low ALT levels were significantly associated with AD diagnosis, increased amyloid PET burden, and faster longitudinal decline in cognitive function in both cohorts. In particular, the AST to ALT ratio was associated with cerebrospinal fluid Aβ42 levels exclusively in the APOE ε4 carrier group in the Alzheimer's Disease Neuroimaging Initiative cohort but not with phosphorylated tau181 or total tau levels. Moreover, mediation analyses from both cohorts revealed that in the APOE ε4 carriers group, age did not mediate the associations between liver enzymes and AD diagnosis or amyloid PET burden. However, amyloid PET burden partially mediated the association between liver enzymes and AD diagnosis exclusively in the APOE ε4 carriers group. Conclusion This study provides valuable insights into the significant association of the APOE ε4 allele with liver enzymes and their potential role in Aβ-related pathogenesis and cognition in AD. Further research is required to elucidate the underlying mechanisms and potential therapeutic implications of these findings.
Collapse
Affiliation(s)
- Sang-Won Han
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Sang-Hwa Lee
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Jong Ho Kim
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Jae-Jun Lee
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jong-Hee Sohn
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
7
|
Pinheiro FI, Araújo-Filho I, do Rego ACM, de Azevedo EP, Cobucci RN, Guzen FP. Hepatopancreatic metabolic disorders and their implications in the development of Alzheimer's disease and vascular dementia. Ageing Res Rev 2024; 96:102250. [PMID: 38417711 DOI: 10.1016/j.arr.2024.102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Dementia has been faced with significant public health challenges and economic burdens that urges the need to develop safe and effective interventions. In recent years, an increasing number of studies have focused on the relationship between dementia and liver and pancreatic metabolic disorders that result in diseases such as diabetes, obesity, hypertension and dyslipidemia. Previous reports have shown that there is a plausible correlation between pathologies caused by hepatopancreatic dysfunctions and dementia. Glucose, insulin and IGF-1 metabolized in the liver and pancreas probably have an important influence on the pathophysiology of the most common dementias: Alzheimer's and vascular dementia. This current review highlights recent studies aimed at identifying convergent mechanisms, such as insulin resistance and other diseases, linked to altered hepatic and pancreatic metabolism, which are capable of causing brain changes that ultimately lead to dementia.
Collapse
Affiliation(s)
- Francisco I Pinheiro
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Irami Araújo-Filho
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Amália C M do Rego
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Eduardo P de Azevedo
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil
| | - Ricardo N Cobucci
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil; Postgraduate Program in Science Applied to Women`s Health, Medical School, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Fausto P Guzen
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil; Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil.
| |
Collapse
|
8
|
Wang Y, Liu W, Sun Y, Dong X. Transthyretin-Penetratin: A Potent Fusion Protein Inhibitor against Alzheimer's Amyloid-β Fibrillogenesis with High Blood Brain Barrier Crossing Capability. Bioconjug Chem 2024; 35:419-431. [PMID: 38450606 DOI: 10.1021/acs.bioconjchem.4c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The design of a potent amyloid-β protein (Aβ) inhibitor plays a pivotal role in the prevention and treatment of Alzheimer's disease (AD). Despite endogenous transthyretin (TTR) being recognized as an Aβ inhibitor, the weak inhibitory and blood brain barrier (BBB) crossing capabilities hinder it for Aβ aggregation inhibition and transport. Therefore, we have herein designed a recombinant TTR by conjugating a cationic cell penetrating peptide (penetratin, Pen), which not only enabled the fusion protein, TTR-Pen (TP), to present high BBB penetration but also greatly enhanced the potency of Aβ inhibition. Namely, the protein fusion made TP positively charged, leading to a potent suppression of Aβ40 fibrillization at a low concentration (1.5 μM), while a TTR concentration as high as 12.5 μM was required to gain a similar function. Moreover, TP could mitigate Aβ-induced neuronal death, increase cultured cell viability from 72% to 92% at 2.5 μM, and extend the lifespan of AD nematodes from 14 to 18 d. Thermodynamic studies revealed that TP, enriched in positive charges, presented extensive electrostatic interactions with Aβ40. Importantly, TP showed excellent BBB penetration performance, with a 10 times higher BBB permeability than TTR, which would allow TP to enter the brain of AD patients and participate in the transport of Aβ species out of the brain. Thus, it is expected that the fusion protein has great potential for drug development in AD treatment.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Wei Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
9
|
Joshi SM, Wilson TC, Li Z, Preshlock S, Gómez-Vallejo V, Gouverneur V, Llop J, Arsequell G. Synthesis and PET Imaging Biodistribution Studies of Radiolabeled Iododiflunisal, a Transthyretin Tetramer Stabilizer, Candidate Drug for Alzheimer's Disease. Molecules 2024; 29:488. [PMID: 38257401 PMCID: PMC10818730 DOI: 10.3390/molecules29020488] [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: 11/26/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The small-molecule iododiflunisal (IDIF) is a transthyretin (TTR) tetramer stabilizer and acts as a chaperone of the TTR-Amyloid beta interaction. Oral administration of IDIF improves Alzheimer's Disease (AD)-like pathology in mice, although the mechanism of action and pharmacokinetics remain unknown. Radiolabeling IDIF with positron or gamma emitters may aid in the in vivo evaluation of IDIF using non-invasive nuclear imaging techniques. In this work, we report an isotopic exchange reaction to obtain IDIF radiolabeled with 18F. [19F/18F]exchange reaction over IDIF in dimethyl sulfoxide at 160 °C resulted in the formation of [18F]IDIF in 7 ± 3% radiochemical yield in a 20 min reaction time, with a final radiochemical purity of >99%. Biodistribution studies after intravenous administration of [18F]IDIF in wild-type mice using positron emission tomography (PET) imaging showed capacity to cross the blood-brain barrier (ca. 1% of injected dose per gram of tissue in the brain at t > 10 min post administration), rapid accumulation in the liver, long circulation time, and progressive elimination via urine. Our results open opportunities for future studies in larger animal species or human subjects.
Collapse
Affiliation(s)
- Sameer M. Joshi
- CIC BiomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Parque Tecnológico de San Sebastián, 20009 Donostia-San Sebastián, Spain; (S.M.J.); (V.G.-V.)
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Thomas C. Wilson
- Chemistry Research Laboratory, Oxford University, Oxford OX1 3TA, UK; (T.C.W.); (V.G.)
| | - Zibo Li
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Sean Preshlock
- Chemistry Research Laboratory, Oxford University, Oxford OX1 3TA, UK; (T.C.W.); (V.G.)
| | - Vanessa Gómez-Vallejo
- CIC BiomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Parque Tecnológico de San Sebastián, 20009 Donostia-San Sebastián, Spain; (S.M.J.); (V.G.-V.)
| | - Véronique Gouverneur
- Chemistry Research Laboratory, Oxford University, Oxford OX1 3TA, UK; (T.C.W.); (V.G.)
| | - Jordi Llop
- CIC BiomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, Parque Tecnológico de San Sebastián, 20009 Donostia-San Sebastián, Spain; (S.M.J.); (V.G.-V.)
| | - Gemma Arsequell
- Institut de Química Avançada de Catalunya (IQAC), Spanish National Research Council (CSIC), 08034 Barcelona, Spain
| |
Collapse
|
10
|
Loeffler DA. Approaches for Increasing Cerebral Efflux of Amyloid-β in Experimental Systems. J Alzheimers Dis 2024; 100:379-411. [PMID: 38875041 PMCID: PMC11307100 DOI: 10.3233/jad-240212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Amyloid protein-β (Aβ) concentrations are increased in the brain in both early onset and late onset Alzheimer's disease (AD). In early onset AD, cerebral Aβ production is increased and its clearance is decreased, while increased Aβ burden in late onset AD is due to impaired clearance. Aβ has been the focus of AD therapeutics since development of the amyloid hypothesis, but efforts to slow AD progression by lowering brain Aβ failed until phase 3 trials with the monoclonal antibodies lecanemab and donanemab. In addition to promoting phagocytic clearance of Aβ, antibodies lower cerebral Aβ by efflux of Aβ-antibody complexes across the capillary endothelia, dissolving Aβ aggregates, and a "peripheral sink" mechanism. Although the blood-brain barrier is the main route by which soluble Aβ leaves the brain (facilitated by low-density lipoprotein receptor-related protein-1 and ATP-binding cassette sub-family B member 1), Aβ can also be removed via the blood-cerebrospinal fluid barrier, glymphatic drainage, and intramural periarterial drainage. This review discusses experimental approaches to increase cerebral Aβ efflux via these mechanisms, clinical applications of these approaches, and findings in clinical trials with these approaches in patients with AD or mild cognitive impairment. Based on negative findings in clinical trials with previous approaches targeting monomeric Aβ, increasing the cerebral efflux of soluble Aβ is unlikely to slow AD progression if used as monotherapy. But if used as an adjunct to treatment with lecanemab or donanemab, this approach might allow greater slowing of AD progression than treatment with either antibody alone.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
11
|
Han SW, Lee SH, Kim JH, Lee JJ, Park YH, Kim S, Nho K, Sohn JH. Association of Serum Liver Enzymes with Brain Amyloidopathy and Cognitive Performance. J Alzheimers Dis Rep 2023; 7:1465-1474. [PMID: 38225965 PMCID: PMC10789293 DOI: 10.3233/adr-230148] [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: 10/19/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024] Open
Abstract
Background Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation and neurofibrillary tangles in the brain. Emerging evidence has suggested potential interactions between the brain and periphery, particularly the liver, in regulating Aβ homeostasis. Objective This study aimed to investigate the association of serum liver enzymes with brain amyloidopathy and cognitive performance in patients with complaints of cognitive decline. Methods A total of 1,036 patients (mean age 74 years, 66.2% female) with subjective cognitive decline, mild cognitive impairment, AD dementia, and other neurodegenerative diseases were included using the Smart Clinical Data Warehouse. Amyloid positron emission tomography (PET) imaging, comprehensive neuropsychological evaluations, and measurements of liver enzymes, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase, total bilirubin, and albumin, were assessed. After propensity score matching, logistic and linear regression analyses were used to investigate the associations between liver enzymes, amyloid status, and cognitive performance. Additionally, a machine learning approach was used to assess the classification performance of liver enzymes in predicting amyloid PET positivity. Results Lower ALT levels and higher AST-to-ALT ratios were significantly associated with amyloid PET positivity and AD diagnosis. The AST-to-ALT ratio was also significantly associated with poor memory function. Machine learning analysis revealed that the classification performance of amyloid status (AUC = 0.642) for age, sex, and apolipoprotein E ɛ4 carrier status significantly improved by 6.2% by integrating the AST-to-ALT ratio. Conclusions These findings highlight the potential association of liver function on AD and its potential as a diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Sang-Won Han
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gangwon-do, Republic of Korea
| | - Sang-Hwa Lee
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gangwon-do, Republic of Korea
| | - Jong Ho Kim
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Jae-Jun Lee
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jong-Hee Sohn
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gangwon-do, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
12
|
Ghosh S, Villacorta-Martin C, Lindstrom-Vautrin J, Kenney D, Golden CS, Edwards CV, Sanchorawala V, Connors LH, Giadone RM, Murphy GJ. Mapping cellular response to destabilized transthyretin reveals cell- and amyloidogenic protein-specific signatures. Amyloid 2023; 30:379-393. [PMID: 37439769 DOI: 10.1080/13506129.2023.2224494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/04/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND In ATTR amyloidosis, transthyretin (TTR) protein is secreted from the liver and deposited as toxic aggregates at downstream target tissues. Despite recent advancements in treatments for ATTR amyloidosis, the mechanisms underlying misfolded TTR-mediated cellular damage remain elusive. METHODS In an effort to define early events of TTR-associated stress, we exposed neuronal (SH-SY5Y) and cardiac (AC16) cells to wild-type and destabilized TTR variants (TTRV122I (p.V142I) and TTRL55P (p.L70P)) and performed transcriptional (RNAseq) and epigenetic (ATACseq) profiling. We subsequently compared TTR-responsive signatures to cells exposed to destabilized antibody light chain protein associated with AL amyloidosis as well as ER stressors (thapsigargin, heat shock). RESULTS In doing so, we observed overlapping, yet distinct cell type- and amyloidogenic protein-specific signatures, suggesting unique responses to each amyloidogenic variant. Moreover, we identified chromatin level changes in AC16 cells exposed to mutant TTR that resolved upon pre-incubation with kinetic stabilizer tafamidis. CONCLUSIONS Collectively, these data provide insight into the mechanisms underlying destabilized protein-mediated cellular damage and provide a robust resource representing cellular responses to aggregation-prone proteins and ER stress.
Collapse
Affiliation(s)
- Sabrina Ghosh
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
| | | | | | - Devin Kenney
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Carly S Golden
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Camille V Edwards
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
- Amyloidosis Center, Alan and Sandra Gerry Amyloid Research Laboratory, Boston University School of Medicine, Boston, MA, USA
| | - Vaishali Sanchorawala
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
- Amyloidosis Center, Alan and Sandra Gerry Amyloid Research Laboratory, Boston University School of Medicine, Boston, MA, USA
| | - Lawreen H Connors
- Amyloidosis Center, Alan and Sandra Gerry Amyloid Research Laboratory, Boston University School of Medicine, Boston, MA, USA
| | - Richard M Giadone
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
| | - George J Murphy
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine, Section of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
13
|
Nakamura M, Imaoka M, Tazaki F, Hida M, Imai R, Kubo T, Sakai K, Takeda M. Serum transthyretin level and its relationship with cognitive function in community-dwelling older people: Cross sectional and longitudinal study. Arch Gerontol Geriatr 2023; 115:105226. [PMID: 37837790 DOI: 10.1016/j.archger.2023.105226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Serum transthyretin (TTR) level has suggested association with mild cognitive impairment (MCI) and dementia. To clarify its usefulness as a biomarker of change in cognitive function in older individuals with normal cognitive function (NC) as a phenotype, we investigated the relationship between cognitive scores and TTR levels. We also investigated the involvement of TTR in the transition from NC to MCI. METHODS Cognitive function was evaluated using Addenbrooke's Cognitive Examination-Revised (ACE-R). A cross-sectional study was conducted in community-dwelling older people (n = 211) with NC, MCI, or dementia according to ACE-R scores. A 32-month longitudinal study was then conducted (n = 29). RESULTS Mean TTR levels did not differ between the NC, MCI and dementia groups. Linear regression analysis showed a significant relationship in people with NC between TTR and ACE-R (β = -0.192; p < 0.001). Multiple regression analysis adjusted for stepwise procedure-selected covariates showed that TTR was significantly associated with ACE-R in people with NC (β = -0.130; p = 0.014). Furthermore, multiple regression analysis showed significant association between TTR level and memory (β = -0.584; p = 0.002) and with language (β = -0.743; p = 0.031) in people with NC. In the longitudinal study, mean TTR level at baseline in women with MCI was significantly higher than that in women with NC (p = 0.044). CONCLUSIONS Serum TTR level is suggested to be associated with cognitive scores in people with NC and to be an indicator of progression from NC to MCI.
Collapse
Affiliation(s)
- Misa Nakamura
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Graduate School of Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan.
| | - Masakazu Imaoka
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Graduate School of Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| | - Fumie Tazaki
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| | - Mitsumasa Hida
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Graduate School of Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| | - Ryota Imai
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Graduate School of Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| | - Takanari Kubo
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| | - Keiko Sakai
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| | - Masatoshi Takeda
- Cognitive Reserve Research Center, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan; Graduate School of Osaka Kawasaki Rehabilitation University, 158 Mizuma, Kaizuka, Osaka 597-0104, Japan
| |
Collapse
|
14
|
Iqbal J, Bibi M, Huang GD, Xue YX, Khatttak JZK, Yang M, Jia XJ. Differential regulation of hippocampal transcriptome by circulating estrogen. Funct Integr Genomics 2023; 23:309. [PMID: 37735249 DOI: 10.1007/s10142-023-01234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Estrogen (E2) modulates the synaptic structure and plasticity in the hippocampus. Previous studies showed that E2 fluctuations during various phases of the menstrual cycle produce subtle neurosynaptic changes that impact women's behavior, emotion, and cognitive functions. In this study, we explored the transcriptome of the hippocampus via RNA-seq (RNA-sequencing) between proestrus (PE) and diestrus (DE) stages in young female rats to determine the effect of E2 of PE and DE stages on hippocampal gene expression. We identified 238 genes (at 1.5-fold-change selection criteria, FDR adjusted p-value < 0.05) as differentially expressed genes (DEGs) that responded to E2 between PE and DE stages. Functional analysis based on Gene Ontology (GO) revealed that a higher E2 level corresponded to an increase in gene transcription among most of the DEGs, suggesting biological mechanisms operating differentially in the hippocampus of female rats between PE and DE stages in the estrus cycle; while analysis with Kyoto Encyclopedia of Genes and Genomes database (KEGG) found that the DEGs involving neuroactive ligand-receptor interaction, antigen processing, cell adhesion molecules, and presentation were upregulated in PE stage, whereas DEGs in pathways relating to bile secretion, coagulation cascades, osteoclast differentiation, cysteine and methionine metabolism were upregulated in DE stage of the estrus cycle. The high-fold expression of DEGs was confirmed by a follow-up quantitative real-time PCR. Our findings in this current study have provided fundamental information for further dissection of neuro-molecular mechanisms in the hippocampus in response to E2 fluctuation and its relationship with disorders.
Collapse
Affiliation(s)
- Javed Iqbal
- Shenzhen Graduate School, Peking University, Shenzhen, China
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No.77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China
| | - Maryam Bibi
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Geng-Di Huang
- Shenzhen Graduate School, Peking University, Shenzhen, China
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No.77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | | | - Mei Yang
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No.77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China.
- Clinical College of Mental Health, Shenzhen University Health Science Center, Shenzhen, China.
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China.
| | - Xiao-Jian Jia
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No.77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China.
- Clinical College of Mental Health, Shenzhen University Health Science Center, Shenzhen, China.
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
15
|
Ciccone L, Camodeca C, Tonali N, Barlettani L, Rossello A, Fruchart Gaillard C, Kaffy J, Petrarolo G, La Motta C, Nencetti S, Orlandini E. New Hybrid Compounds Incorporating Natural Products as Multifunctional Agents against Alzheimer's Disease. Pharmaceutics 2023; 15:2369. [PMID: 37896129 PMCID: PMC10610016 DOI: 10.3390/pharmaceutics15102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 10/29/2023] Open
Abstract
A series of new hybrid derivatives 1a-c, 2a-c, 3a-c, 4a-c, 5a-c, inspired by nature, were synthesized and studied as multifunctional agents for the treatment of Alzheimer's disease (AD). These compounds were designed to merge together the trifluoromethyl benzyloxyaminic bioactive moiety, previously identified, with different acids available in nature. The ability of the synthesized compounds to chelate biometals, such as Cu2+, Zn2+ and Fe2+, was studied by UV-Vis spectrometer, and through a preliminary screening their antioxidant activity was evaluated by DPPH. Then, selected compounds were tested by in vitro ABTS free radical method and ex vivo rat brain TBARS assay. Compounds 2a-c, combining the strongest antioxidant and biometal chelators activities, were studied for their ability to contrast Aβ1-40 fibrillization process. Finally, starting from the promising profile obtained for compound 2a, we evaluated if it could be able to induce a positive cross-interaction between transthyretin (TTR) and Aβ in presence and in absence of Cu2+.
Collapse
Affiliation(s)
- Lidia Ciccone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
| | - Caterina Camodeca
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
| | - Nicolò Tonali
- CNRS, BioCIS, Bâtiment Henri Moissan, Université Paris-Saclay, 17 Av. des Sciences, 91400 Orsay, France; (N.T.); (J.K.)
| | - Lucia Barlettani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
| | - Armando Rossello
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy;
| | - Carole Fruchart Gaillard
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Université Paris Saclay, 91191 Gif-sur-Yvette, France;
| | - Julia Kaffy
- CNRS, BioCIS, Bâtiment Henri Moissan, Université Paris-Saclay, 17 Av. des Sciences, 91400 Orsay, France; (N.T.); (J.K.)
| | - Giovanni Petrarolo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
| | - Concettina La Motta
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
| | - Susanna Nencetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (C.C.); (L.B.); (A.R.); (G.P.); (C.L.M.)
| | - Elisabetta Orlandini
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy;
- Department of Earth Sciences, University of Pisa, Via Santa Maria 53, 56126 Pisa, Italy
| |
Collapse
|
16
|
Ullah R, Lee EJ. Advances in Amyloid-β Clearance in the Brain and Periphery: Implications for Neurodegenerative Diseases. Exp Neurobiol 2023; 32:216-246. [PMID: 37749925 PMCID: PMC10569141 DOI: 10.5607/en23014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
This review examines the role of impaired amyloid-β clearance in the accumulation of amyloid-β in the brain and the periphery, which is closely associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). The molecular mechanism underlying amyloid-β accumulation is largely unknown, but recent evidence suggests that impaired amyloid-β clearance plays a critical role in its accumulation. The review provides an overview of recent research and proposes strategies for efficient amyloid-β clearance in both the brain and periphery. The clearance of amyloid-β can occur through enzymatic or non-enzymatic pathways in the brain, including neuronal and glial cells, blood-brain barrier, interstitial fluid bulk flow, perivascular drainage, and cerebrospinal fluid absorption-mediated pathways. In the periphery, various mechanisms, including peripheral organs, immunomodulation/immune cells, enzymes, amyloid-β-binding proteins, and amyloid-β-binding cells, are involved in amyloid-β clearance. Although recent findings have shed light on amyloid-β clearance in both regions, opportunities remain in areas where limited data is available. Therefore, future strategies that enhance amyloid-β clearance in the brain and/or periphery, either through central or peripheral clearance approaches or in combination, are highly encouraged. These strategies will provide new insight into the disease pathogenesis at the molecular level and explore new targets for inhibiting amyloid-β deposition, which is central to the pathogenesis of sporadic AD (amyloid-β in parenchyma) and CAA (amyloid-β in blood vessels).
Collapse
Affiliation(s)
- Rahat Ullah
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Neurology, School of Medicine, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
17
|
Gullotta GS, Costantino G, Sortino MA, Spampinato SF. Microglia and the Blood-Brain Barrier: An External Player in Acute and Chronic Neuroinflammatory Conditions. Int J Mol Sci 2023; 24:ijms24119144. [PMID: 37298096 DOI: 10.3390/ijms24119144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system that guarantee immune surveillance and exert also a modulating role on neuronal synaptic development and function. Upon injury, microglia get activated and modify their morphology acquiring an ameboid phenotype and pro- or anti-inflammatory features. The active role of microglia in blood-brain barrier (BBB) function and their interaction with different cellular components of the BBB-endothelial cells, astrocytes and pericytes-are described. Here, we report the specific crosstalk of microglia with all the BBB cell types focusing in particular on the involvement of microglia in the modulation of BBB function in neuroinflammatory conditions that occur in conjunction with an acute event, such as a stroke, or in a slow neurodegenerative disease, such as Alzheimer's disease. The potential of microglia to exert a dual role, either protective or detrimental, depending on disease stages and environmental conditioning factors is also discussed.
Collapse
Affiliation(s)
- Giorgia Serena Gullotta
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuseppe Costantino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Ph.D. Program in Neuroscience and Education, DISTUM, University of Foggia, 71121 Foggia, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | | |
Collapse
|
18
|
Tekin E, Aslan Karakelle N, Dinçer S. Effects of taurine on metal cations, transthyretin and LRP-1 in a rat model of Alzheimer's disease. J Trace Elem Med Biol 2023; 79:127219. [PMID: 37229981 DOI: 10.1016/j.jtemb.2023.127219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Researches on diagnosis and treatment of Alzheimer's disease, the most common type of dementia, are still ongoing. Taurine is frequently used in Alzheimer's disease models due to its protective effects. Metal cation dyshomeostasis is an important etiological factor for Alzheimer's disease. Transthyretin protein is thought to act as a transporter for the Aβ protein that accumulates in the brain and is eliminated in the liver and kidneys via the LRP-1 receptor. However, the effect of taurine on this mechanisms is not fully known. METHODS 30 male rats, aged 28 ± 4 months, were divided into 5 groups (n = 6) as follows: control group, sham group, Aβ 1-42 group, taurine group and taurine+Aβ 1-42 group. Oral taurine pre-supplementation was given as 1000 mg/kg-body weight/day for 6 weeks to taurine and taurine+Aβ 1-42 groups. RESULTS Plasma copper, heart transthyretin and Aβ 1-42, brain and kidney LRP-1 levels were found to be decreased in the Aβ 1-42 group. Brain transthyretin was higher in taurine+Aβ 1-42 group and brain Aβ 1-42 was higher in Aβ 1-42 and taurine+Aβ 1-42 groups. CONCLUSION Taurine pre-supplementation maintained cardiac transthyretin levels, decreased cardiac Aβ 1-42 levels and increased brain and kidney LRP-1 levels. Taurine may have a potential to be used as a protective agent for aged people at high risk for Alzheimer's disease.
Collapse
Affiliation(s)
- Esra Tekin
- Gazi University Faculty of Medicine, Department of Physiology, Ankara, Turkey.
| | - Nida Aslan Karakelle
- Lokman Hekim University Faculty of Medicine, Department of Physiology, Ankara, Turkey.
| | - Sibel Dinçer
- Gazi University Faculty of Medicine, Department of Physiology, Ankara, Turkey.
| |
Collapse
|
19
|
da Luz MHM, Pino JMV, Mônico-Neto M, de Amorim PN, Antunes HKM, Porcionatto MA, Lee KS. Sleep deprivation modulates APOE and LDL receptor-related protein 1 through thyroid hormone T4 and impairs Aβ clearance in hippocampus of rats. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166729. [PMID: 37137431 DOI: 10.1016/j.bbadis.2023.166729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Alzheimer's disease is the most common form of dementia. One of its pathological hallmarks is Aβ accumulation, which is influenced by APOE genotype and expression, as well as by sleep homeostasis. However, conflicting mechanisms for APOE roles in Aβ clearance have been reported, and the relationship between APOE and sleep also remains unclear. In this study, we aimed to investigate how hormonal alteration caused by sleep deprivation affects APOE and its receptors in rats, and to evaluate the role of different cell types in Aβ clearance. Paradoxical sleep deprivation for 96 h increased Aβ level in hippocampus with concomitant reduction of APOE and LRP1 at the time point within the resting period. Sleep deprivation also significantly reduced T4 levels in both active and resting times. To evaluate the effect of T4 variation, C6 glial cells and primary brain endothelial cells were treated with T4. High T4 level (300 ng/mL) increased APOE, but reduced LRP1 and LDL-R in C6 cells, while in primary endothelial cells, LDL-R levels were increased. Treatment of C6 cells with exogenous APOE reduced LRP1 and Aβ uptake. These results suggest that T4 modulates LRP1 and LDL-R in both cell types, but in the opposite manner, thus, sleep deprivation might modify the ratio of the receptors in blood-brain barrier and glial cells by altering T4 levels. Considering that LRP1 and LDL-R are important for Aβ clearance, sleep deprivation might also affect the degree of participation of glia in Aβ clearance, and consequently, turnover of Aβ in the brain.
Collapse
Affiliation(s)
| | | | - Marcos Mônico-Neto
- Post Graduate Program of Interdisciplinary Health Sciences, Universidade Federal de São Paulo, Santos, Brazil
| | | | | | | | - Kil Sun Lee
- Department of Biochemistry, Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
20
|
Maity S, Mayer MG, Shu Q, Linh H, Bao D, Blair RV, He Y, Lyon CJ, Hu TY, Fischer T, Fan J. Cerebrospinal Fluid Protein Markers Indicate Neuro-Damage in SARS-CoV-2-Infected Nonhuman Primates. Mol Cell Proteomics 2023; 22:100523. [PMID: 36870567 PMCID: PMC9981268 DOI: 10.1016/j.mcpro.2023.100523] [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: 07/13/2022] [Revised: 02/18/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Neurologic manifestations are among the most frequently reported complications of COVID-19. However, given the paucity of tissue samples and the highly infectious nature of the etiologic agent of COVID-19, we have limited information to understand the neuropathogenesis of COVID-19. Therefore, to better understand the impact of COVID-19 on the brain, we used mass-spectrometry-based proteomics with a data-independent acquisition mode to investigate cerebrospinal fluid (CSF) proteins collected from two different nonhuman primates, Rhesus Macaque and African Green Monkeys, for the neurologic effects of the infection. These monkeys exhibited minimal to mild pulmonary pathology but moderate to severe central nervous system (CNS) pathology. Our results indicated that CSF proteome changes after infection resolution corresponded with bronchial virus abundance during early infection and revealed substantial differences between the infected nonhuman primates and their age-matched uninfected controls, suggesting these differences could reflect altered secretion of CNS factors in response to SARS-CoV-2-induced neuropathology. We also observed the infected animals exhibited highly scattered data distributions compared to their corresponding controls indicating the heterogeneity of the CSF proteome change and the host response to the viral infection. Dysregulated CSF proteins were preferentially enriched in functional pathways associated with progressive neurodegenerative disorders, hemostasis, and innate immune responses that could influence neuroinflammatory responses following COVID-19. Mapping these dysregulated proteins to the Human Brain Protein Atlas found that they tended to be enriched in brain regions that exhibit more frequent injury following COVID-19. It, therefore, appears reasonable to speculate that such CSF protein changes could serve as signatures for neurologic injury, identify important regulatory pathways in this process, and potentially reveal therapeutic targets to prevent or attenuate the development of neurologic injuries following COVID-19.
Collapse
Affiliation(s)
- Sudipa Maity
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Meredith G Mayer
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA
| | - Qingbo Shu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Hellmers Linh
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA
| | - Duran Bao
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Robert V Blair
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Christopher J Lyon
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tony Y Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tracy Fischer
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| |
Collapse
|
21
|
Lee SB, Yu J, Kim H, Kim KW, Jeong JW, Kim YL, Park SJ, Koo TS, Lee C, Hong KB, Choi S. Novel Strategy To Inhibit Transthyretin Amyloidosis via the Synergetic Effect of Chemoselective Acylation and Noncovalent Inhibitor Release. J Med Chem 2023; 66:2893-2903. [PMID: 36749109 DOI: 10.1021/acs.jmedchem.2c01926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Strategies for developing targeted covalent inhibitors (TCIs), which have the advantages of a prolonged duration of action and selectivity toward a drug target, have attracted great interest in drug discovery. Herein, we report chemoselective covalent inhibitors that specifically target lysine ε-amine groups that conjugate with an endogenous protein to prevent disease-causing protein misfolding and aggregation. These TCIs are unique because the benzoyl group is preferentially conjugated to Lys15 at the top of the T4 binding site within transthyretin (TTR) while simultaneously releasing a potent noncovalent TTR kinetic stabilizer. The potency of these covalent inhibitors is superior to tafamidis, the only FDA-approved drug for the treatment of hereditary TTR amyloidosis. In addition to investigations into the covalent modification of TTR via reverse-phase high-performance liquid chromatography, direct methods are performed to confirm and visualize the presumed covalent interaction via mass spectrometry and X-ray crystallography.
Collapse
Affiliation(s)
- Seok Beom Lee
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| | - Jaeni Yu
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| | - Hyunwoo Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Kun Woo Kim
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| | - Jong Woo Jeong
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| | - Yun Lan Kim
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| | - Sung Jean Park
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| | - Tae-Sung Koo
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| | - Changwook Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ki Bum Hong
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Sungwook Choi
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejon 34134, Republic of Korea
| |
Collapse
|
22
|
Nezvedová M, Jha D, Váňová T, Gadara D, Klímová H, Raška J, Opálka L, Bohačiaková D, Spáčil Z. Single Cerebral Organoid Mass Spectrometry of Cell-Specific Protein and Glycosphingolipid Traits. Anal Chem 2023; 95:3160-3167. [PMID: 36724094 PMCID: PMC10016744 DOI: 10.1021/acs.analchem.2c00981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cerebral organoids are a prolific research topic and an emerging model system for neurological diseases in human neurobiology. However, the batch-to-batch reproducibility of current cultivation protocols is challenging and thus requires a high-throughput methodology to comprehensively characterize cerebral organoid cytoarchitecture and neural development. We report a mass spectrometry-based protocol to quantify neural tissue cell markers, cell surface lipids, and housekeeping proteins in a single organoid. Profiled traits probe the development of neural stem cells, radial glial cells, neurons, and astrocytes. We assessed the cell population heterogeneity in individually profiled organoids in the early and late neurogenesis stages. Here, we present a unifying view of cell-type specificity of profiled protein and lipid traits in neural tissue. Our workflow characterizes the cytoarchitecture, differentiation stage, and batch cultivation variation on an individual cerebral organoid level.
Collapse
Affiliation(s)
- Markéta Nezvedová
- RECETOX, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
| | - Durga Jha
- RECETOX, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
| | - Tereza Váňová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno 625 00, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno 656 91, Czech Republic
| | - Darshak Gadara
- RECETOX, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
| | - Hana Klímová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno 625 00, Czech Republic
| | - Jan Raška
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno 625 00, Czech Republic
| | - Lukáš Opálka
- Department of Chemistry, Faculty of Pharmacy, Charles University, Hradec Králové 500 05, Czech Republic
| | - Dáša Bohačiaková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno 625 00, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno 656 91, Czech Republic
| | - Zdeněk Spáčil
- RECETOX, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
| |
Collapse
|
23
|
Omega-3 PUFAs as a Dietary Supplement in Senile Systemic Amyloidosis. Nutrients 2023; 15:nu15030749. [PMID: 36771455 PMCID: PMC9921273 DOI: 10.3390/nu15030749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Eicosapentaenoic acid (EPA; 20:5) and docosahexaenoic acid (DHA; 22:6), two omega-3 poly-unsaturated fatty acids (PUFAs), are the main components in oil derived from fish and other marine organisms. EPA and DHA are commercially available as dietary supplements and are considered to be very safe and contribute to guaranteeing human health. Studies report that PUFAs have a role in contrasting neurodegenerative processes related to amyloidogenic proteins, such as β-amyloid for AD, α-synuclein in PD, and transthyretin (TTR) in TTR amyloidosis. In this context, we investigated if EPA and DHA can interact directly with TTR, binding inside the thyroxin-binding pockets (T4BP) that contribute to the tetramer stabilization. The data obtained showed that EPA and DHA can contribute to stabilizing the TTR tetramer through interactions with T4BP.
Collapse
|
24
|
Koopmans F, Li KW, Klaassen RV, Smit AB. MS-DAP Platform for Downstream Data Analysis of Label-Free Proteomics Uncovers Optimal Workflows in Benchmark Data Sets and Increased Sensitivity in Analysis of Alzheimer's Biomarker Data. J Proteome Res 2022; 22:374-386. [PMID: 36541440 PMCID: PMC9903323 DOI: 10.1021/acs.jproteome.2c00513] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the rapidly moving proteomics field, a diverse patchwork of data analysis pipelines and algorithms for data normalization and differential expression analysis is used by the community. We generated a mass spectrometry downstream analysis pipeline (MS-DAP) that integrates both popular and recently developed algorithms for normalization and statistical analyses. Additional algorithms can be easily added in the future as plugins. MS-DAP is open-source and facilitates transparent and reproducible proteome science by generating extensive data visualizations and quality reporting, provided as standardized PDF reports. Second, we performed a systematic evaluation of methods for normalization and statistical analysis on a large variety of data sets, including additional data generated in this study, which revealed key differences. Commonly used approaches for differential testing based on moderated t-statistics were consistently outperformed by more recent statistical models, all integrated in MS-DAP. Third, we introduced a novel normalization algorithm that rescues deficiencies observed in commonly used normalization methods. Finally, we used the MS-DAP platform to reanalyze a recently published large-scale proteomics data set of CSF from AD patients. This revealed increased sensitivity, resulting in additional significant target proteins which improved overlap with results reported in related studies and includes a large set of new potential AD biomarkers in addition to previously reported.
Collapse
Affiliation(s)
- Frank Koopmans
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands,
| | - Ka Wan Li
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands
| | - Remco V. Klaassen
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands
| | - August B. Smit
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
25
|
Saavedra J, Nascimento M, Liz MA, Cardoso I. Key brain cell interactions and contributions to the pathogenesis of Alzheimer's disease. Front Cell Dev Biol 2022; 10:1036123. [PMID: 36523504 PMCID: PMC9745159 DOI: 10.3389/fcell.2022.1036123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/14/2022] [Indexed: 06/22/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide, with the two major hallmarks being the deposition of extracellular β-amyloid (Aβ) plaques and of intracellular neurofibrillary tangles (NFTs). Additionally, early pathological events such as cerebrovascular alterations, a compromised blood-brain barrier (BBB) integrity, neuroinflammation and synaptic dysfunction, culminate in neuron loss and cognitive deficits. AD symptoms reflect a loss of neuronal circuit integrity in the brain; however, neurons do not operate in isolation. An exclusively neurocentric approach is insufficient to understand this disease, and the contribution of other brain cells including astrocytes, microglia, and vascular cells must be integrated in the context. The delicate balance of interactions between these cells, required for healthy brain function, is disrupted during disease. To design successful therapies, it is critical to understand the complex brain cellular connections in AD and the temporal sequence of their disturbance. In this review, we discuss the interactions between different brain cells, from physiological conditions to their pathological reactions in AD, and how this basic knowledge can be crucial for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Joana Saavedra
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Mariana Nascimento
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Márcia A. Liz
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Isabel Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Anusha-Kiran Y, Mol P, Dey G, Bhat FA, Chatterjee O, Deolankar SC, Philip M, Prasad TSK, Srinivas Bharath MM, Mahadevan A. Regional heterogeneity in mitochondrial function underlies region specific vulnerability in human brain ageing: Implications for neurodegeneration. Free Radic Biol Med 2022; 193:34-57. [PMID: 36195160 DOI: 10.1016/j.freeradbiomed.2022.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/22/2022] [Indexed: 12/01/2022]
Abstract
Selective neuronal vulnerability (SNV) of specific neuroanatomical regions such as frontal cortex (FC) and hippocampus (HC) is characteristic of age-associated neurodegenerative diseases (NDDs), although its pathogenetic basis remains unresolved. We hypothesized that physiological differences in mitochondrial function in neuroanatomical regions could contribute to SNV. To investigate this, we evaluated mitochondrial function in human brains (age range:1-90 y) in FC, striatum (ST), HC, cerebellum (CB) and medulla oblongata (MD), using enzyme assays and quantitative proteomics. Striking differences were noted in resistant regions- MD and CB compared to the vulnerable regions- FC, HC and ST. At younger age (25 ± 5 y), higher activity of electron transport chain enzymes and upregulation of metabolic and antioxidant proteins were noted in MD compared to FC and HC, that was sustained with increasing age (≥65 y). In contrast, the expression of synaptic proteins was higher in FC, HC and ST (vs. MD). In line with this, quantitative phospho-proteomics revealed activation of upstream regulators (ERS, PPARα) of mitochondrial metabolism and inhibition of synaptic pathways in MD. Microtubule Associated Protein Tau (MAPT) showed overexpression in FC, HC and ST both in young and older age (vs. MD). MAPT hyperphosphorylation and the activation of its kinases were noted in FC and HC with age. Our study demonstrates that regional heterogeneity in mitochondrial and other cellular functions contribute to SNV and protect regions such as MD, while rendering FC and HC vulnerable to NDDs. The findings also support the "last in, first out" hypothesis of ageing, wherein regions such as FC, that are the most recent to develop phylogenetically and ontogenetically, are the first to be affected in ageing and NDDs.
Collapse
Affiliation(s)
- Yarlagadda Anusha-Kiran
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, 560029, India; Department of Clinical Psychopharmacology and Neurotoxicology, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India
| | - Praseeda Mol
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Gourav Dey
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India
| | - Firdous Ahmad Bhat
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India
| | - Oishi Chatterjee
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Sayali Chandrashekhar Deolankar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Mariamma Philip
- Department of Biostatistics, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India.
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, 560029, India.
| |
Collapse
|
27
|
Wang C, Zeng L, Li Y, Shi C, Peng Y, Pan R, Huang M, Wang S, Zhang J, Li H. Decabromodiphenyl ethane induces locomotion neurotoxicity and potential Alzheimer's disease risks through intensifying amyloid-beta deposition by inhibiting transthyretin/transthyretin-like proteins. ENVIRONMENT INTERNATIONAL 2022; 168:107482. [PMID: 35998411 DOI: 10.1016/j.envint.2022.107482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
As a major alternative to traditional brominated flame retardants (BFRs), decabromodiphenyl ethane (DBDPE) is widely used and has been commonly detected in various environmental media and organisms. Few previous studies have focused on DBDPE-induced locomotion neurotoxicity, and the exact molecular mechanisms and related health risks remain unclear. In this study, we first analyzed the locomotion indicators of nematodes following DBDPE exposure, demonstrated that DBDPE caused locomotion neurotoxicity, and identified that a series of the transthyretin (TTR)-like genes participated in the regulation of nematode motility by transcriptomic analysis, gene transcription validation and TTR-like mutant verification. Subsequently, this study demonstrated that DBDPE exacerbated amyloid-beta (Aβ) deposition by repressing TTR/TTR-like gene transcription based on Alzheimer's disease (AD) model nematodes and human SH-SY5Y cells following DBDPE exposure and further revealed that DBDPE reduced the binding between TTR and Aβ by competing with the strand G region sites on the TTR/TTR-like protein, ultimately exacerbating Aβ deposition and the risk of AD. In short, our study demonstrated that DBDPE induced locomotion neurotoxicity and potential AD risks through intensifying Aβ deposition by inhibiting TTR/TTR-like proteins, providing reference support for risk management and policy formulation related to DBDPE and similarly structured novel BFRs.
Collapse
Affiliation(s)
- Chen Wang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Lingjun Zeng
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Yeyong Li
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Chongli Shi
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Yi Peng
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Ruolin Pan
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Mengyan Huang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Susu Wang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Jin Zhang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China
| | - Hui Li
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
28
|
Antioxidant Quercetin 3-O-Glycosylated Plant Flavonols Contribute to Transthyretin Stabilization. CRYSTALS 2022. [DOI: 10.3390/cryst12050638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Plants are rich in secondary metabolites, which are often useful as a relevant source of nutraceuticals. Quercetin (QUE) is a flavonol aglycone able to bind Transthyretin (TTR), a plasma protein that under pathological conditions can lose its native structure leading to fibrils formation and amyloid diseases onset. Here, the dual nature of five quercetin 3-O-glycosylated flavonol derivatives, isolated from different plant species, such as possible binders of TTR and antioxidants, was investigated. The crystal structure of 3-O-β-D-galactopyranoside in complex with TTR was solved, suggesting that not only quercetin but also its metabolites can contribute to stabilizing the TTR tetramer.
Collapse
|
29
|
Uchida K. Waste Clearance in the Brain and Neuroinflammation: A Novel Perspective on Biomarker and Drug Target Discovery in Alzheimer's Disease. Cells 2022; 11:cells11050919. [PMID: 35269541 PMCID: PMC8909773 DOI: 10.3390/cells11050919] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/26/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial disease with a heterogeneous etiology. The pathology of Alzheimer’s disease is characterized by amyloid-beta and hyperphosphorylated tau, which are necessary for disease progression. Many clinical trials on disease-modifying drugs for AD have failed to indicate their clinical benefits. Recent advances in fundamental research have indicated that neuroinflammation plays an important pathological role in AD. Damage- and pathogen-associated molecular patterns in the brain induce neuroinflammation and inflammasome activation, causing caspase-1-dependent glial and neuronal cell death. These waste products in the brain are eliminated by the glymphatic system via perivascular spaces, the blood-brain barrier, and the blood–cerebrospinal fluid barrier. Age-related vascular dysfunction is associated with an impairment of clearance and barrier functions, leading to neuroinflammation. The proteins involved in waste clearance in the brain and peripheral circulation may be potential biomarkers and drug targets in the early stages of cognitive impairment. This short review focuses on waste clearance dysfunction in AD pathobiology and discusses the improvement of waste clearance as an early intervention in prodromal AD and preclinical stages of dementia.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba 305-8575, Ibaraki, Japan; ; Tel.: +81-29-853-3210; Fax: +81-50-3730-7456
- Institute for Biomedical Research, MCBI, 4-9-29 Matsushiro, Tsukuba 305-0035, Ibaraki, Japan
| |
Collapse
|
30
|
Shippy DC, Watters JJ, Ulland TK. Transcriptional response of murine microglia in Alzheimer’s disease and inflammation. BMC Genomics 2022; 23:183. [PMID: 35247975 PMCID: PMC8898509 DOI: 10.1186/s12864-022-08417-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Background Alzheimer’s disease (AD) is a neurodegenerative disorder and is the most common cause of late-onset dementia. Microglia, the primary innate immune cells of the central nervous system (CNS), have a complex role in AD neuropathology. In the initial stages of AD, microglia play a role in limiting pathology by removing amyloid-β (Aβ) by phagocytosis. In contrast, microglia also release pro-inflammatory cytokines and chemokines to promote neuroinflammation and exacerbate AD neuropathology. Therefore, investigating microglial gene networks could identify new targets for therapeutic strategies for AD. Results We identified 465 differentially expressed genes (DEG) in 5XFAD versus wild-type mice by microarray, 354 DEG in lipopolysaccharide (LPS)-stimulated N9 microglia versus unstimulated control cells using RNA-sequencing (RNA-seq), with 32 DEG common between both datasets. Analyses of the 32 common DEG uncovered numerous molecular functions and pathways involved in Aβ phagocytosis and neuroinflammation associated with AD. Furthermore, multiplex ELISA confirmed the induction of several cytokines and chemokines in LPS-stimulated microglia. Conclusions In summary, AD triggered multiple signaling pathways that regulate numerous genes in microglia, contributing to Aβ phagocytosis and neuroinflammation. Overall, these data identified several regulatory factors and biomarkers in microglia that could be useful in further understanding AD neuropathology. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08417-8.
Collapse
|
31
|
Identifying new molecular players in extracellular proteostasis. Biochem Soc Trans 2021; 50:321-334. [PMID: 34940856 DOI: 10.1042/bst20210369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 02/02/2023]
Abstract
Proteostasis refers to a delicately tuned balance between the processes of protein synthesis, folding, localization, and the degradation of proteins found inside and outside cells. Our understanding of extracellular proteostasis is rather limited and largely restricted to knowledge of 11 currently established extracellular chaperones (ECs). This review will briefly outline what is known of the established ECs, before moving on to discuss experimental strategies used to identify new members of this growing family, and an examination of a group of putative new ECs identified using one of these approaches. An observation that emerges from an analysis of the expanding number of ECs is that all of these proteins are multifunctional. Strikingly, the armory of activities each possess uniquely suit them as a group to act together at sites of tissue damage, infection, and inflammation to restore homeostasis. Lastly, we highlight outstanding questions to guide future research in this field.
Collapse
|
32
|
Cotrina EY, Santos LM, Rivas J, Blasi D, Leite JP, Liz MA, Busquets MA, Planas A, Prohens R, Gimeno A, Jiménez-Barbero J, Gales L, Llop J, Quintana J, Cardoso I, Arsequell G. Targeting transthyretin in Alzheimer's disease: Drug discovery of small-molecule chaperones as disease-modifying drug candidates for Alzheimer's disease. Eur J Med Chem 2021; 226:113847. [PMID: 34555615 DOI: 10.1016/j.ejmech.2021.113847] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022]
Abstract
Transthyretin (TTR) has a well-established role in neuroprotection in Alzheimer's Disease (AD). We have setup a drug discovery program of small-molecule compounds that act as chaperones enhancing TTR/Amyloid-beta peptide (Aβ) interactions. A combination of computational drug repurposing approaches and in vitro biological assays have resulted in a set of molecules which were then screened with our in-house validated high-throughput screening ternary test. A prioritized list of chaperones was obtained and corroborated with ITC studies. Small-molecule chaperones have been discovered, among them our lead compound Iododiflunisal (IDIF), a molecule in the discovery phase; one investigational drug (luteolin); and 3 marketed drugs (sulindac, olsalazine and flufenamic), which could be directly repurposed or repositioned for clinical use. Not all TTR tetramer stabilizers behave as chaperones in vitro. These chemically diverse chaperones will be used for validating TTR as a target in vivo, and to select one repurposed drug as a candidate to enter clinical trials as AD disease-modifying drug.
Collapse
Affiliation(s)
- Ellen Y Cotrina
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), E-08034, Barcelona, Spain
| | - Luis Miguel Santos
- IBMC - Instituto de Biologia Molecular e Celular, PT-4200-135, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PT-4200-135, Porto, Portugal
| | - Josep Rivas
- Plataforma Drug Discovery, Parc Científic de Barcelona (PCB), E-08028, Barcelona, Spain
| | - Daniel Blasi
- Plataforma Drug Discovery, Parc Científic de Barcelona (PCB), E-08028, Barcelona, Spain
| | - José Pedro Leite
- IBMC - Instituto de Biologia Molecular e Celular, PT-4200-135, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PT-4200-135, Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), PT-4050-013, Porto, Portugal
| | - Márcia A Liz
- IBMC - Instituto de Biologia Molecular e Celular, PT-4200-135, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PT-4200-135, Porto, Portugal
| | - Maria Antònia Busquets
- Facultat de Farmàcia i Ciències de l'Alimentació, University of Barcelona, E-08028, Barcelona, Spain
| | - Antoni Planas
- Institut Químic de Sarrià, Universitat Ramon Llull, E-08017, Barcelona, Spain
| | - Rafel Prohens
- Centres Científics i Tecnologics, Universitat de Barcelona, E-08028, Barcelona, Spain
| | - Ana Gimeno
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, E-48160, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, E-48160, Derio, Spain; Ikerbasque, Basque Foundation for Science, E-48009, Bilbao, Spain
| | - Luis Gales
- IBMC - Instituto de Biologia Molecular e Celular, PT-4200-135, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PT-4200-135, Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), PT-4050-013, Porto, Portugal
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), E-20014, San Sebastian, Spain
| | - Jordi Quintana
- Plataforma Drug Discovery, Parc Científic de Barcelona (PCB), E-08028, Barcelona, Spain.
| | - Isabel Cardoso
- IBMC - Instituto de Biologia Molecular e Celular, PT-4200-135, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PT-4200-135, Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), PT-4050-013, Porto, Portugal.
| | - Gemma Arsequell
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), E-08034, Barcelona, Spain.
| |
Collapse
|
33
|
Araghi M, Shipley MJ, Anand A, Mills NL, Kivimaki M, Singh-Manoux A, Tabák A, Sabia S, Brunner EJ. Serum transthyretin and risk of cognitive decline and dementia: 22-year longitudinal study. Neurol Sci 2021; 42:5093-5100. [PMID: 33770310 PMCID: PMC9136660 DOI: 10.1007/s10072-021-05191-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022]
Abstract
Serum transthyretin (TTR) may be an early biomarker for Alzheimer's disease and related disorders (ADRD). We investigated associations of TTR measured at baseline with cognitive decline and incident ADRD and whether TTR trajectories differ between ADRD cases and non-cases, over 22 years before diagnosis. A total of 6024 adults aged 45-69 in 1997-1999 were followed up until 2019. TTR was assessed three times, and 297 cases of dementia were recorded. Higher TTR was associated with higher cognitive function at baseline; however, TTR was unrelated to subsequent change in cognitive function. TTR at baseline did not predict ADRD risk (hazard ratio per SD TTR (4.8 mg/dL) = 0.97; 95% confidence interval: 0.94-1.00). Among those later diagnosed with ADRD, there was a marginally steeper downward TTR trajectory than those free of ADRD over follow-up (P=0.050). Our findings suggest TTR is not neuroprotective. The relative decline in TTR level in the preclinical stage of ADRD is likely to be a consequence of disease processes.
Collapse
Affiliation(s)
- Marzieh Araghi
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK.
| | - Martin J Shipley
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Atul Anand
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Archana Singh-Manoux
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
- Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Adam Tabák
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
- Department of Internal Medicine and Oncology, Semmelweis University Faculty of Medicine, Budapest, Hungary
- Department of Public Health, Semmelweis University Faculty of Medicine, Budapest, Hungary
| | - Séverine Sabia
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
- Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Eric J Brunner
- Department of Epidemiology and Public Health, Institute of Epidemiology and Health Care, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| |
Collapse
|
34
|
Exploring Molecular Mechanisms Involved in the Development of the Depression-Like Phenotype in Interleukin-18-Deficient Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9975865. [PMID: 34708129 PMCID: PMC8545524 DOI: 10.1155/2021/9975865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/18/2021] [Indexed: 11/18/2022]
Abstract
Interleukin-18 (IL-18) is an inflammatory cytokine that has been linked to energy homeostasis and psychiatric symptoms such as depression and cognitive impairment. We previously revealed that deficiency in IL-18 led to hippocampal abnormalities and resulted in depression-like symptoms. However, the impact of IL-18 deficiency on other brain regions remains to be clarified. In this study, we first sought to confirm that IL-18 expression in neural cells can be found in human brain tissue. Subsequently, we examined the expression of genes in the prefrontal cortex of Il18−/− mice and compared it with gene expression in mice subjected to a chronic mild stress model of depression. Extracted genes were further analyzed using Ingenuity® Pathway Analysis, in which 18 genes common to both the chronic mild stressed model and Il18−/− mice were identified. Of those, 16 were significantly differentially expressed between Il18+/+ and Il18−/− mice. We additionally measured protein expression of α-2-HS-glycoprotein (AHSG) and transthyretin (TTR) in serum and the brain. In the prefrontal cortex of Il18−/− mice, TTR but not AHSG was significantly decreased. Conversely, in the serum of Il18−/− mice, AHSG was significantly increased but not TTR. Therefore, our results suggest that in IL-18-deficit conditions, TTR in the brain is one of the mediators causally related to depression, and AHSG in peripheral organs is one of the regulators inducing energy imbalance. Moreover, this study suggests a possible “signpost” to clarify the molecular mechanisms commonly underlying the immune system, energy metabolism, neural function, and depressive disorders.
Collapse
|
35
|
Peng HB, de Lannoy IAM, Pang KS. Measuring Amyloid-β Peptide Concentrations in Murine Brain with Improved ELISA Assay. Curr Protoc 2021; 1:e253. [PMID: 34661993 DOI: 10.1002/cpz1.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The amyloid-β (Aβ) peptides of 40 and 42 amino acids that are implicated in Alzheimer's disease may potentially aggregate into toxic oligomers and form neuritic plaques. The enzyme-linked immunosorbent assay (ELISA) is a facile method used for the determination of Aβ concentrations in biological matrices, namely plasma, cerebrospinal fluid, and brain. The method is mostly used for the measurement of Aβ concentrations in transgenic mice, but it is unknown whether the ELISA method is suitable for measuring low, endogenous levels of Aβ in the brains of wild-type mice. The Aβ ELISA kit manufacturer recommends use of 5 M guanidine hydrochloride (GuHCl), a protein-denaturing agent, for homogenization of the brain tissue, followed by dilution back down to 0.1 M to avoid quenching by GuHCl. Components of brain matrices and GuHCl that could interfere with the quantitation have not been investigated. In this article, we describe an improved method involving homogenization of mouse brain with 1 M instead of 5 M GuHCl, reducing the dilution factor by 5× to provide a higher sensitivity. The modified ELISA assay is improved for the quantitation of brain Aβ peptides in wild-type mice, where Aβ peptide levels are much lower than those in transgenic mouse models. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- H Benson Peng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | - K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Rejc L, Gómez-Vallejo V, Rios X, Cossío U, Baz Z, Mujica E, Gião T, Cotrina EY, Jiménez-Barbero J, Quintana J, Arsequell G, Cardoso I, Llop J. Oral Treatment with Iododiflunisal Delays Hippocampal Amyloid-β Formation in a Transgenic Mouse Model of Alzheimer's Disease: A Longitudinal in vivo Molecular Imaging Study1. J Alzheimers Dis 2021; 77:99-112. [PMID: 32804152 DOI: 10.3233/jad-200570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Transthyretin (TTR) is a tetrameric, amyloid-β (Aβ)-binding protein, which reduces Aβ toxicity. The TTR/Aβ interaction can be enhanced by a series of small molecules that stabilize its tetrameric form. Hence, TTR stabilizers might act as disease-modifying drugs in Alzheimer's disease. OBJECTIVE We monitored the therapeutic efficacy of two TTR stabilizers, iododiflunisal (IDIF), which acts as small-molecule chaperone of the TTR/Aβ interaction, and tolcapone, which does not behave as a small-molecule chaperone, in an animal model of Alzheimer's disease using positron emission tomography (PET). METHODS Female mice (AβPPswe/PS1A246E/TTR+/-) were divided into 3 groups (n = 7 per group): IDIF-treated, tolcapone-treated, and non-treated. The oral treatment (100 mg/Kg/day) was started at 5 months of age. Treatment efficacy assessment was based on changes in longitudinal deposition of Aβ in the hippocampus (HIP) and the cortex (CTX) and determined using PET-[18F]florbetaben. Immunohistochemical analysis was performed at age = 14 months. RESULTS Standard uptake values relative to the cerebellum (SUVr) of [18F]florbetaben in CTX and HIP of non-treated animals progressively increased from age = 5 to 11 months and stabilized afterwards. In contrast, [18F]florbetaben uptake in HIP of IDIF-treated animals remained constant between ages = 5 and 11 months and significantly increased at 14 months. In the tolcapone-treated group, SUVr progressively increased with time, but at lower rate than in the non-treated group. No significant treatment effect was observed in CTX. Results from immunohistochemistry matched the in vivo data at age = 14 months. CONCLUSION Our work provides encouraging preliminary results on the ability of small-molecule chaperones to ameliorate Aβ deposition in certain brain regions.
Collapse
Affiliation(s)
- Luka Rejc
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Ljubljana, Slovenia
| | - Vanessa Gómez-Vallejo
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastián, Guipúzcoa, Spain
| | - Xabier Rios
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastián, Guipúzcoa, Spain
| | - Unai Cossío
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastián, Guipúzcoa, Spain
| | - Zuriñe Baz
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastián, Guipúzcoa, Spain
| | - Edurne Mujica
- Biochemistry and Molecular Biology, EHU-UPV, Leioa, Spain
| | - Tiago Gião
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ellen Y Cotrina
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), Barcelona, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Bizkaia, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.,Department Organic Chemistry II, Faculty Science & Technology, EHU-UPV, Leioa, Spain
| | - Jordi Quintana
- Plataforma Drug Discovery, Parc Científic de Barcelona (PCB), Barcelona, Spain
| | - Gemma Arsequell
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), Barcelona, Spain
| | - Isabel Cardoso
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastián, Guipúzcoa, Spain.,Centro de Investigación Biomédica en Red - Enfermedades Respiratorias (CIBERES)
| |
Collapse
|
37
|
Magalhães J, Eira J, Liz MA. The role of transthyretin in cell biology: impact on human pathophysiology. Cell Mol Life Sci 2021; 78:6105-6117. [PMID: 34297165 PMCID: PMC11073172 DOI: 10.1007/s00018-021-03899-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/08/2021] [Accepted: 07/09/2021] [Indexed: 01/29/2023]
Abstract
Transthyretin (TTR) is an extracellular protein mainly produced in the liver and choroid plexus, with a well-stablished role in the transport of thyroxin and retinol throughout the body and brain. TTR is prone to aggregation, as both wild-type and mutated forms of the protein can lead to the accumulation of amyloid deposits, resulting in a disease called TTR amyloidosis. Recently, novel activities for TTR in cell biology have emerged, ranging from neuronal health preservation in both central and peripheral nervous systems, to cellular fate determination, regulation of proliferation and metabolism. Here, we review the novel literature regarding TTR new cellular effects. We pinpoint TTR as major player on brain health and nerve biology, activities that might impact on nervous systems pathologies, and assign a new link between TTR and angiogenesis and cancer. We also explore the molecular mechanisms underlying TTR activities at the cellular level, and suggest that these might go beyond its most acknowledged carrier functions and include interaction with receptors and activation of intracellular signaling pathways.
Collapse
Affiliation(s)
- Joana Magalhães
- Neurodegeneration Team, Nerve Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Jessica Eira
- Neurodegeneration Team, Nerve Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade Do Porto, Porto, Portugal
| | - Márcia Almeida Liz
- Neurodegeneration Team, Nerve Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
38
|
Gião T, Saavedra J, Vieira JR, Pinto MT, Arsequell G, Cardoso I. Neuroprotection in early stages of Alzheimer's disease is promoted by transthyretin angiogenic properties. ALZHEIMERS RESEARCH & THERAPY 2021; 13:143. [PMID: 34429155 PMCID: PMC8385857 DOI: 10.1186/s13195-021-00883-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Background While still controversial, it has been demonstrated that vascular defects can precede the onset of other AD hallmarks features, making it an important therapeutic target. Given that the protein transthyretin (TTR) has been established as neuroprotective in AD, here we investigated the influence of TTR in the vasculature. Methods We evaluated the thickness of the basement membrane and the length of brain microvessels, by immunohistochemistry, in AβPPswe/PS1A246E (AD) transgenic mice and non-transgenic mice (NT) bearing one (TTR+/−) or two (TTR+/+) copies of the TTR gene. The angiogenic potential of TTR was evaluated in vitro using the tube formation assay, and in vivo using the chick chorioallantoic membrane (CAM) assay. Results AD transgenic mice with TTR genetic reduction, AD/TTR+/−, exhibited a thicker BM in brain microvessels and decreased vessel length than animals with normal TTR levels, AD/TTR+/+. Further in vivo investigation, using the CAM assay, revealed that TTR is a pro-angiogenic molecule, and the neovessels formed are functional. Also, TTR increased the expression of key angiogenic molecules such as proteins interleukins 6 and 8, angiopoietin 2, and vascular endothelial growth factor, by endothelial cells, in vitro, under tube formation conditions. We showed that while TTR reduction also leads to a thicker BM in NT mice, this effect is more pronounced in AD mice than in NT animals, strengthening the idea that TTR is a neuroprotective protein. We also studied the effect of TTR tetrameric stabilization on BM thickness, showing that AD mice treated with the TTR tetrameric stabilizer iododiflunisal (IDIF) displayed a significant reduction of BM thickness and increased vessel length, when compared to non-treated littermates. Conclusion Our in vivo results demonstrate the involvement of TTR in angiogenesis, particularly as a modulator of vascular alterations occurring in AD. Since TTR is decreased early in AD, its tetrameric stabilization can represent a therapeutic avenue for the early treatment of AD through the maintenance of the vascular structure. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00883-8.
Collapse
Affiliation(s)
- Tiago Gião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), 4050-013, Porto, Portugal
| | - Joana Saavedra
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - José Ricardo Vieira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.,Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marta Teixeira Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.,IPATIMUP - Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho,45-, 4200-135, Porto, Portugal
| | - Gemma Arsequell
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034, Barcelona, Spain
| | - Isabel Cardoso
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal. .,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), 4050-013, Porto, Portugal.
| |
Collapse
|
39
|
Ge F, Zhu D, Tian M, Shi J. The Role of Thyroid Function in Alzheimer's Disease. J Alzheimers Dis 2021; 83:1553-1562. [PMID: 34420955 DOI: 10.3233/jad-210339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The thyroid gland is crucial for the regulation of metabolism, growth, and development of various tissues, organs, systems, including the central nervous system. Recent studies have implicated the role of thyroid dysfunction in the etiology of Alzheimer's disease (AD), while AD leads to a significant increase in the prevalence of thyroid dysfunction. In this review, we have analyzed the role of thyroid function in the pathophysiology of AD as well as its biomarkers. The present review aims to provide encouraging targets for early screening of AD risk factors and intervention strategies.
Collapse
Affiliation(s)
- Feifei Ge
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Donglin Zhu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Minjie Tian
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jingping Shi
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Wieczorek E, Ożyhar A. Transthyretin: From Structural Stability to Osteoarticular and Cardiovascular Diseases. Cells 2021; 10:1768. [PMID: 34359938 PMCID: PMC8307983 DOI: 10.3390/cells10071768] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Accepted: 07/09/2021] [Indexed: 01/10/2023] Open
Abstract
Transthyretin (TTR) is a tetrameric protein transporting hormones in the plasma and brain, which has many other activities that have not been fully acknowledged. TTR is a positive indicator of nutrition status and is negatively correlated with inflammation. TTR is a neuroprotective and oxidative-stress-suppressing factor. The TTR structure is destabilized by mutations, oxidative modifications, aging, proteolysis, and metal cations, including Ca2+. Destabilized TTR molecules form amyloid deposits, resulting in senile and familial amyloidopathies. This review links structural stability of TTR with the environmental factors, particularly oxidative stress and Ca2+, and the processes involved in the pathogenesis of TTR-related diseases. The roles of TTR in biomineralization, calcification, and osteoarticular and cardiovascular diseases are broadly discussed. The association of TTR-related diseases and vascular and ligament tissue calcification with TTR levels and TTR structure is presented. It is indicated that unaggregated TTR and TTR amyloid are bound by vicious cycles, and that TTR may have an as yet undetermined role(s) at the crossroads of calcification, blood coagulation, and immune response.
Collapse
Affiliation(s)
- Elżbieta Wieczorek
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland;
| | | |
Collapse
|
41
|
Breaking the barrier: In vitro models to study choroid plexus development. Curr Opin Cell Biol 2021; 73:41-49. [PMID: 34182208 DOI: 10.1016/j.ceb.2021.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022]
Abstract
The choroid plexus is central to normal brain function by secreting cerebrospinal fluid and dynamically regulating its composition throughout development and homoeostasis. Much of our current understanding of this region of the brain comes from studies in animal models. These fundamental studies have shed light on choroid plexus mechanisms of secretion, barrier function and homoeostatic regulation. However, how these specific mechanisms are regulated in the human choroid plexus is much less understood, due to ethical and technical limitations. A number of recent breakthroughs have enabled a new range of techniques and tools for functional characterisation of choroid plexus development and physiology. With the advance of new technologies such as in vivo imaging, single-cell transcriptomics and in vitro three-dimensional cultures we are now able to address a number of outstanding questions in choroid plexus biology. Here, we discuss some of these recent breakthroughs and we focus in particular on how in vitro models can be a powerful tool to study human cerebrospinal fluid secretion and barrier function.
Collapse
|
42
|
Yang Q, Li AP. Messenger RNA Expression of Albumin, Transferrin, Transthyretin, Asialoglycoprotein Receptor, Cytochrome P450 Isoform, Uptake Transporter, and Efflux Transporter Genes as a Function of Culture Duration in Prolonged Cultured Cryopreserved Human Hepatocytes as Collagen-Matrigel Sandwich Cultures: Evidence for Redifferentiation upon Prolonged Culturing. Drug Metab Dispos 2021; 49:790-802. [PMID: 34135090 DOI: 10.1124/dmd.121.000424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/10/2021] [Indexed: 01/04/2023] Open
Abstract
Hepatic gene expression as a function of culture duration was evaluated in prolonged cultured human hepatocytes. Human hepatocytes from seven donors were maintained as near-confluent collagen-Matrigelsandwich cultures, with messenger RNA expression for genes responsible for key hepatic functions quantified by real-time polymerase chain reaction at culture durations of 0 (day of plating), 2, 7, 9, 16, 23, 26, 29, 36, and 43 days. Key hepatocyte genes were evaluated, including the differentiation markers albumin, transferrin, and transthyretin; the hepatocyte-specific asialoglycoprotein receptor 1 cytochrome P450 isoforms CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A7; uptake transporter isoforms SLC10A1, SLC22A1, SLC22A7, SLCO1B1, SLCO1B3, and SLCO2B1; efflux transporter isoforms ATP binding cassette (ABC)B1, ABCB11, ABCC2, ABCC3, ABCC4, and ABCG2; and the nonspecific housekeeping gene hypoxanthine ribosyl transferase 1 (HPRT1). The well established dedifferentiation phenomenon was observed on day 2, with substantial (>80%) decreases in gene expression in day 2 cultures observed for all genes evaluated except HPRT1 and efflux transporters ABCB1, ABCC2, ABCC3 (<50% decrease in expression), ABCC4 (>400% increase in expression), and ABCG2 (no decrease in expression). All genes with a >80% decrease in expression were found to have increased levels of expression on day 7, with peak expression observed on either day 7 or day 9, followed by a gradual decrease in expression up to the longest duration evaluated of 43 days. Our results provide evidence that cultured human hepatocytes undergo redifferentiation upon prolonged culturing. SIGNIFICANCE STATEMENT: This study reports that although human hepatocytes underwent dedifferentiation upon 2 days of culture, prolonged culturing resulted in redifferentiation based on gene expression of differentiation markers, uptake and efflux transporters, and cytochrome P450 isoforms. The observed redifferentiation suggests that prolonged (>7 days) culturing of human hepatocyte cultures may represent an experimental approach to overcome the initial dedifferentiation process, resulting in "stabilized" hepatocytes that can be applied toward the evaluation of drug properties requiring an extended period of treatment and evaluation.
Collapse
Affiliation(s)
- Qian Yang
- In Vitro ADMET Laboratories Inc., Columbia, Maryland
| | - Albert P Li
- In Vitro ADMET Laboratories Inc., Columbia, Maryland
| |
Collapse
|
43
|
Exploring the Physiological Role of Transthyretin in Glucose Metabolism in the Liver. Int J Mol Sci 2021; 22:ijms22116073. [PMID: 34199897 PMCID: PMC8200108 DOI: 10.3390/ijms22116073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022] Open
Abstract
Transthyretin (TTR), a 55 kDa evolutionarily conserved protein, presents altered levels in several conditions, including malnutrition, inflammation, diabetes, and Alzheimer’s Disease. It has been shown that TTR is involved in several functions, such as insulin release from pancreatic β-cells, recovery of blood glucose and glucagon levels of the islets of Langerhans, food intake, and body weight. Here, the role of TTR in hepatic glucose metabolism was explored by studying the levels of glucose in mice with different TTR genetic backgrounds, namely with two copies of the TTR gene, TTR+/+; with only one copy, TTR+/−; and without TTR, TTR−/−. Results showed that TTR haploinsufficiency (TTR+/−) leads to higher glucose in both plasma and in primary hepatocyte culture media and lower expression of the influx glucose transporters, GLUT1, GLUT3, and GLUT4. Further, we showed that TTR haploinsufficiency decreases pyruvate kinase M type (PKM) levels in mice livers, by qRT-PCR, but it does not affect the hepatic production of the studied metabolites, as determined by 1H NMR. Finally, we demonstrated that TTR increases mitochondrial density in HepG2 cells and that TTR insufficiency triggers a higher degree of oxidative phosphorylation in the liver. Altogether, these results indicate that TTR contributes to the homeostasis of glucose by regulating the levels of glucose transporters and PKM enzyme and by protecting against mitochondrial oxidative stress.
Collapse
|
44
|
Schwab N, Ju Y, Hazrati LN. Early onset senescence and cognitive impairment in a murine model of repeated mTBI. Acta Neuropathol Commun 2021; 9:82. [PMID: 33964983 PMCID: PMC8106230 DOI: 10.1186/s40478-021-01190-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (mTBI) results in broad neurological symptoms and an increased risk of being diagnosed with a neurodegenerative disease later in life. While the immediate oxidative stress response and post-mortem pathology of the injured brain has been well studied, it remains unclear how early pathogenic changes may drive persistent symptoms and confer susceptibility to neurodegeneration. In this study we have used a mouse model of repeated mTBI (rmTBI) to identify early gene expression changes at 24 h or 7 days post-injury (7 dpi). At 24 h post-injury, gene expression of rmTBI mice shows activation of the DNA damage response (DDR) towards double strand DNA breaks, altered calcium and cell–cell signalling, and inhibition of cell death pathways. By 7 dpi, rmTBI mice had a gene expression signature consistent with induction of cellular senescence, activation of neurodegenerative processes, and inhibition of the DDR. At both timepoints gliosis, microgliosis, and axonal damage were evident in the absence of any gross lesion, and by 7 dpi rmTBI also mice had elevated levels of IL1β, p21, 53BP1, DNA2, and p53, supportive of DNA damage-induced cellular senescence. These gene expression changes reflect establishment of processes usually linked to brain aging and suggests that cellular senescence occurs early and most likely prior to the accumulation of toxic proteins. These molecular changes were accompanied by spatial learning and memory deficits in the Morris water maze. To conclude, we have identified DNA damage-induced cellular senescence as a repercussion of repeated mild traumatic brain injury which correlates with cognitive impairment. Pathways involved in senescence may represent viable treatment targets of post-concussive syndrome. Senescence has been proposed to promote neurodegeneration and appears as an effective target to prevent long-term complications of mTBI, such as chronic traumatic encephalopathy and other related neurodegenerative pathologies.
Collapse
|
45
|
Soares Martins T, Marçalo R, Ferreira M, Vaz M, Silva RM, Martins Rosa I, Vogelgsang J, Wiltfang J, da Cruz e Silva OAB, Henriques AG. Exosomal Aβ-Binding Proteins Identified by "In Silico" Analysis Represent Putative Blood-Derived Biomarker Candidates for Alzheimer´s Disease. Int J Mol Sci 2021; 22:ijms22083933. [PMID: 33920336 PMCID: PMC8070602 DOI: 10.3390/ijms22083933] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
The potential of exosomes as biomarker resources for diagnostics and even for therapeutics has intensified research in the field, including in the context of Alzheimer´s disease (AD). The search for disease biomarkers in peripheral biofluids is advancing mainly due to the easy access it offers. In the study presented here, emphasis was given to the bioinformatic identification of putative exosomal candidates for AD. The exosomal proteomes of cerebrospinal fluid (CSF), serum and plasma, were obtained from three databases (ExoCarta, EVpedia and Vesiclepedia), and complemented with additional exosomal proteins already associated with AD but not found in the databases. The final biofluids’ proteomes were submitted to gene ontology (GO) enrichment analysis and the exosomal Aβ-binding proteins that can constitute putative candidates were identified. Among these candidates, gelsolin, a protein known to be involved in inhibiting Abeta fibril formation, was identified, and it was tested in human samples. The levels of this Aβ-binding protein, with anti-amyloidogenic properties, were assessed in serum-derived exosomes isolated from controls and individuals with dementia, including AD cases, and revealed altered expression patterns. Identification of potential peripheral biomarker candidates for AD may be useful, not only for early disease diagnosis but also in drug trials and to monitor disease progression, allowing for a timely therapeutic intervention, which will positively impact the patient’s quality of life.
Collapse
Affiliation(s)
- Tânia Soares Martins
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Rui Marçalo
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Maria Ferreira
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Margarida Vaz
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Raquel M. Silva
- Center for Interdisciplinary Research in Health (CIIS), Faculdade de Medicina Dentária, Universidade Católica Portuguesa, Estrada da Circunvalação, 3504-505 Viseu, Portugal;
| | - Ilka Martins Rosa
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Von-Siebold-Str. 5, 37075 Goettingen, Germany;
- Translational Neuroscience Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Jens Wiltfang
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Von-Siebold-Str. 5, 37075 Goettingen, Germany;
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Goettingen, Germany
| | - Odete A. B. da Cruz e Silva
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
- Correspondence:
| |
Collapse
|
46
|
Physiological Metals Can Induce Conformational Changes in Transthyretin Structure: Neuroprotection or Misfolding Induction? CRYSTALS 2021. [DOI: 10.3390/cryst11040354] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Transthyretin (TTR) is a plasma homotetrameric protein that transports thyroxine and retinol. TTR itself, under pathological conditions, dissociates into partially unfolded monomers that aggregate and form fibrils. Metal ions such as Zn2+, Cu2+, Fe2+, Mn2+ and Ca2+ play a controversial role in the TTR amyloidogenic pathway. TTR is also present in cerebrospinal fluid (CSF), where it behaves as one of the major Aβ-binding-proteins. The interaction between TTR and Aβ is stronger in the presence of high concentrations of Cu2+. Crystals of TTR, soaked in solutions of physiological metals such as Cu2+ and Fe2+, but not Mn2+, Zn2+, Fe3+, Al3+, Ni2+, revealed an unusual conformational change. Here, we investigate the effects that physiological metals have on TTR, in order to understand if metals can induce a specific and active conformation of TTR that guides its Aβ-scavenging role. The capability of certain metals to induce and accelerate its amyloidogenic process is also discussed.
Collapse
|
47
|
Zhou J, Jangili P, Son S, Ji MS, Won M, Kim JS. Fluorescent Diagnostic Probes in Neurodegenerative Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001945. [PMID: 32902000 DOI: 10.1002/adma.202001945] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/19/2020] [Indexed: 05/22/2023]
Abstract
Neurodegenerative diseases are debilitating disorders that feature progressive and selective loss of function or structure of anatomically or physiologically associated neuronal systems. Both chronic and acute neurodegenerative diseases are associated with high morbidity and mortality along with the death of neurons in different areas of the brain; moreover, there are few or no effective curative therapy options for treating these disorders. There is an urgent need to diagnose neurodegenerative disease as early as possible, and to distinguish between different disorders with overlapping symptoms that will help to decide the best clinical treatment. Recently, in neurodegenerative disease research, fluorescent-probe-mediated biomarker visualization techniques have been gaining increasing attention for the early diagnosis of neurodegenerative diseases. A survey of fluorescent probes for sensing and imaging biomarkers of neurodegenerative diseases is provided. These imaging probes are categorized based on the different potential biomarkers of various neurodegenerative diseases, and their advantages and disadvantages are discussed. Guides to develop new sensing strategies, recognition mechanisms, as well as the ideal features to further improve neurodegenerative disease fluorescence imaging are also explored.
Collapse
Affiliation(s)
- Jin Zhou
- College of Pharmacy, Weifang Medical University, Weifang, 261053, China
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Paramesh Jangili
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Subin Son
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Myung Sun Ji
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Miae Won
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
48
|
Saponaro F, Kim JH, Chiellini G. Transthyretin Stabilization: An Emerging Strategy for the Treatment of Alzheimer's Disease? Int J Mol Sci 2020; 21:ijms21228672. [PMID: 33212973 PMCID: PMC7698513 DOI: 10.3390/ijms21228672] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/14/2020] [Accepted: 11/15/2020] [Indexed: 12/27/2022] Open
Abstract
Transthyretin (TTR), previously named prealbumin is a plasma protein secreted mainly by the liver and choroid plexus (CP) that is a carrier for thyroid hormones (THs) and retinol (vitamin A). The structure of TTR, with four monomers rich in β-chains in a globular tetrameric protein, accounts for the predisposition of the protein to aggregate in fibrils, leading to a rare and severe disease, namely transthyretin amyloidosis (ATTR). Much effort has been made and still is required to find new therapeutic compounds that can stabilize TTR ("kinetic stabilization") and prevent the amyloid genetic process. Moreover, TTR is an interesting therapeutic target for neurodegenerative diseases due to its recognized neuroprotective properties in the cognitive impairment context and interestingly in Alzheimer's disease (AD). Much evidence has been collected regarding the neuroprotective effects in AD, including through in vitro and in vivo studies as well as a wide range of clinical series. Despite this supported hypothesis of neuroprotection for TTR, the mechanisms are still not completely clear. The aim of this review is to highlight the most relevant findings on the neuroprotective role of TTR, and to summarize the recent progress on the development of TTR tetramer stabilizers.
Collapse
Affiliation(s)
| | - Jin Hae Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
| | - Grazia Chiellini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy;
- Correspondence:
| |
Collapse
|
49
|
Sinha A, Chang JC, Xu P, Gindinova K, Cho Y, Sun W, Wu X, Li YM, Greengard P, Kelly JW, Sinha SC. Brain Permeable Tafamidis Amide Analogs for Stabilizing TTR and Reducing APP Cleavage. ACS Med Chem Lett 2020; 11:1973-1979. [PMID: 33062181 PMCID: PMC7549266 DOI: 10.1021/acsmedchemlett.9b00688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/02/2020] [Indexed: 02/08/2023] Open
Abstract
Tafamidis, 1, a potent transthyretin kinetic stabilizer, weakly inhibits the γ-secretase enzyme in vitro. We have synthesized four amide derivatives of 1. These compounds reduce production of the Aβ peptide in N2a695 cells but do not inhibit the γ-secretase enzyme in cell-free assays. By performing fluorescence correlation spectroscopy, we have shown that TTR inhibits Aβ oligomerization and that addition of tafamidis or its amide derivative does not affect TTR's ability to inhibit Aβ oligomerization. The piperazine amide derivative of tafamidis (1a) efficiently penetrates and accumulates in mouse brain and undergoes proteolysis under physiological conditions in mice to produce tafamidis.
Collapse
Affiliation(s)
- Anjana Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Jerry C Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Peng Xu
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Katherina Gindinova
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Younhee Cho
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Weilin Sun
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Xianzhong Wu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Yue Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Subhash C Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| |
Collapse
|
50
|
Vancamp P, Butruille L, Demeneix BA, Remaud S. Thyroid Hormone and Neural Stem Cells: Repair Potential Following Brain and Spinal Cord Injury. Front Neurosci 2020; 14:875. [PMID: 32982671 PMCID: PMC7479247 DOI: 10.3389/fnins.2020.00875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuronal and/or glial cell loss, while traumatic injury is often accompanied by the acute loss of both. Multipotent neural stem cells (NSCs) in the adult mammalian brain spontaneously proliferate, forming neuronal and glial progenitors that migrate toward lesion sites upon injury. However, they fail to replace neurons and glial cells due to molecular inhibition and the lack of pro-regenerative cues. A major challenge in regenerative biology therefore is to unveil signaling pathways that could override molecular brakes and boost endogenous repair. In physiological conditions, thyroid hormone (TH) acts on NSC commitment in the subventricular zone, and the subgranular zone, the two largest NSC niches in mammals, including humans. Here, we discuss whether TH could have beneficial actions in various pathological contexts too, by evaluating recent data obtained in mammalian models of multiple sclerosis (MS; loss of oligodendroglial cells), Alzheimer’s disease (loss of neuronal cells), stroke and spinal cord injury (neuroglial cell loss). So far, TH has shown promising effects as a stimulator of remyelination in MS models, while its role in NSC-mediated repair in other diseases remains elusive. Disentangling the spatiotemporal aspects of the injury-driven repair response as well as the molecular and cellular mechanisms by which TH acts, could unveil new ways to further exploit its pro-regenerative potential, while TH (ant)agonists with cell type-specific action could provide safer and more target-directed approaches that translate easier to clinical settings.
Collapse
Affiliation(s)
- Pieter Vancamp
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Lucile Butruille
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Barbara A Demeneix
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Sylvie Remaud
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| |
Collapse
|