1
|
Tan AYS, Martinez LCMG, Murray HC, Mehrabi NF, Tippett LJ, Turner CP, Curtis MA, Faull RLM, Dragunow M, Singh-Bains MK. Elucidating cortical neurovascular involvement in Huntington's disease using human brain tissue microarrays. Neurobiol Dis 2025:106829. [PMID: 39909082 DOI: 10.1016/j.nbd.2025.106829] [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: 12/09/2024] [Revised: 01/21/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025] Open
Abstract
Although the genetic basis of Huntington's disease (HD) has been determined, the underlying pathophysiological mechanisms contributing to neurodegeneration remain largely unknown. In recent years, increasing evidence has posited vascular dysfunction as a significant early event in disease pathogenesis; however, these processes remain to be fully elucidated. High-content immunohistochemical screening studies were conducted on HD middle temporal gyrus (MTG) human brain tissue microarrays (TMAs) to investigate various components of the vascular system, including endothelial cells (UEA-1), pericytes (PDGFRβ), vascular smooth muscle cells (αSMA), extracellular matrix components (ECM; collagen IV and fibronectin), and leakage markers (haemoglobin and fibrinogen). Analyses of vascular markers revealed an increase in the number of vessels in the HD TMA cohort which was associated with advancing striatal pathology and earlier symptom onset. Furthermore, our findings highlight the preservation of pericytes, vascular smooth muscle cells, ECM components, and blood-brain barrier integrity in the HD MTG. Collectively, the TMA findings allude to mild vascular remodelling in the temporal cortex which is known to present with a lesser degree of neuronal degeneration in HD.
Collapse
Affiliation(s)
- Adelie Y S Tan
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Lance C M G Martinez
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Helen C Murray
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Nasim F Mehrabi
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland 1023, New Zealand
| | - Lynette J Tippett
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; School of Psychology, University of Auckland, Auckland 1023, New Zealand
| | - Clinton P Turner
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland 1023, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland 1023, New Zealand.
| | - Malvindar K Singh-Bains
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland 1023, New Zealand.
| |
Collapse
|
2
|
Qiu J, Peng S, Qu R, Wu L, Xing L, Zhang L, Sun J. New evidence of vascular defects in neurodegenerative diseases revealed by single cell RNA sequencing. Clin Sci (Lond) 2024; 138:1377-1394. [PMID: 39469930 DOI: 10.1042/cs20241658] [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: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Neurodegenerative diseases (NDs) involve the progressive loss of neuronal structure or function in the brain and spinal cord. Despite their diverse etiologies, NDs manifest similar pathologies. Emerging research identifies vascular defects as a previously neglected hallmark of NDs. The development and popularization of single-cell RNA sequencing (scRNA-seq) technologies have significantly advanced our understanding of brain vascular cell types and their molecular characteristics, including gene expression changes at the single-cell level in NDs. These unprecedented insights deepen our understanding of the pathogenic mechanisms underlying NDs. However, the occurrence and role of vascular defects in disease progression remain largely unexplored. In this paper, we systematically summarize recent advances in the structure and organization of the central nervous system vasculature in mice, healthy individuals, and patients with NDs, focussing primarily on disease-specific alterations in vascular cell types or subtypes. Combining scRNA-seq with pathology evidence, we propose that vascular defects, characterized by disruptions in cell types and structural integrity, may serve as common early features of NDs. Finally, we discuss several pathways through which vascular defects in NDs lead to neuronal degeneration. A deeper understanding of the causes and contributions of vascular defects to NDs aids in elucidating the pathogenic mechanisms and developing meaningful therapeutic interventions.
Collapse
Affiliation(s)
- Jiaying Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
3
|
Vasilkovska T, Verschuuren M, Pustina D, van den Berg M, Van Audekerke J, Pintelon I, Cachope R, De Vos WH, Van der Linden A, Adhikari MH, Verhoye M. Evolution of aberrant brain-wide spatiotemporal dynamics of resting-state networks in a Huntington's disease mouse model. Clin Transl Med 2024; 14:e70055. [PMID: 39422700 PMCID: PMC11488302 DOI: 10.1002/ctm2.70055] [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] [Revised: 08/15/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is marked by irreversible loss of neuronal function for which currently no availability for disease-modifying treatment exists. Advances in the understanding of disease progression can aid biomarker development, which in turn can accelerate therapeutic discovery. METHODS We characterised the progression of altered dynamics of whole-brain network states in the zQ175DN mouse model of HD using a dynamic functional connectivity (FC) approach to resting-state fMRI and identified quasi-periodic patterns (QPPs) of brain activity constituting the most prominent resting-state networks. RESULTS The occurrence of the normative QPPs, as observed in healthy controls, was reduced in the HD model as the phenotype progressed. This uncovered progressive cessation of synchronous brain activity with phenotypic progression, which is not observed with the conventional static FC approaches. To better understand the potential underlying cause of the observed changes in these brain states, we further assessed how mutant huntingtin (mHTT) protein deposition affects astrocytes and pericytes - one of the most important effectors of neurovascular coupling, along phenotypic progression. Increased cell-type dependent mHTT deposition was observed at the age of onset of motor anomalies, in the caudate putamen, somatosensory and motor cortex, regions that are prominently involved in HD pathology as seen in humans. CONCLUSION Our findings provide meaningful insights into the development and progression of altered functional brain dynamics in this HD model and open new avenues in assessing the dynamics of whole brain states, through QPPs, in clinical HD research. HIGHLIGHTS Hyperactivity in the LCN-linked regions within short QPPs observed before motor impairment onset. DMLN QPP presents a progressive decrease in DMLN activity and occurrence along HD-like phenotype development. Breakdown of the LCN DMLN state flux at motor onset leads to a subsequent absence of the LCN DMLN QPP at an advanced HD-like stage.
Collapse
Affiliation(s)
- Tamara Vasilkovska
- Bio‐Imaging LabUniversity of AntwerpWilrijkAntwerpBelgium
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Marlies Verschuuren
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
- Laboratory of Cell Biology and HistologyUniversity of AntwerpWilrijkAntwerpBelgium
- Antwerp Centre for Advanced MicroscopyUniversity of AntwerpWilrijkAntwerpBelgium
| | - Dorian Pustina
- CHDI Management, Inc. for CHDI Foundation, Inc.PrincetonNew JerseyUSA
| | - Monica van den Berg
- Bio‐Imaging LabUniversity of AntwerpWilrijkAntwerpBelgium
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Johan Van Audekerke
- Bio‐Imaging LabUniversity of AntwerpWilrijkAntwerpBelgium
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Isabel Pintelon
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
- Laboratory of Cell Biology and HistologyUniversity of AntwerpWilrijkAntwerpBelgium
- Antwerp Centre for Advanced MicroscopyUniversity of AntwerpWilrijkAntwerpBelgium
| | - Roger Cachope
- CHDI Management, Inc. for CHDI Foundation, Inc.PrincetonNew JerseyUSA
| | - Winnok H. De Vos
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
- Laboratory of Cell Biology and HistologyUniversity of AntwerpWilrijkAntwerpBelgium
- Antwerp Centre for Advanced MicroscopyUniversity of AntwerpWilrijkAntwerpBelgium
| | - Annemie Van der Linden
- Bio‐Imaging LabUniversity of AntwerpWilrijkAntwerpBelgium
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Mohit H. Adhikari
- Bio‐Imaging LabUniversity of AntwerpWilrijkAntwerpBelgium
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| | - Marleen Verhoye
- Bio‐Imaging LabUniversity of AntwerpWilrijkAntwerpBelgium
- µNEURO Research Centre of ExcellenceUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
4
|
Mehanna R, Jankovic J. Systemic Symptoms in Huntington's Disease: A Comprehensive Review. Mov Disord Clin Pract 2024; 11:453-464. [PMID: 38529740 PMCID: PMC11078495 DOI: 10.1002/mdc3.14029] [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: 12/02/2023] [Revised: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Although Huntington's disease (HD) is usually thought of as a triad of motor, cognitive, and psychiatric symptoms, there is growing appreciation of HD as a systemic illness affecting the entire body. OBJECTIVES This review aims to draw attention to these systemic non-motor symptoms in HD. METHODS We identified relevant studies published in English by searching MEDLINE (from 1966 to September 2023), using the following subject headings: Huntington disease, autonomic, systemic, cardiovascular, respiratory, gastrointestinal, urinary, sexual and cutaneous, and additional specific symptoms. RESULTS Data from 123 articles were critically reviewed with focus on systemic features associated with HD, such as cardiovascular, respiratory, gastrointestinal, urinary, sexual and sweating. CONCLUSION This systematic review draws attention to a variety of systemic and autonomic co-morbidities in patients with HD. Not all of them correlate with the severity of the primary HD symptoms or CAG repeats. More research is needed to better understand the pathophysiology and treatment of systemic and autonomic dysfunction in HD.
Collapse
Affiliation(s)
- Raja Mehanna
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Vasilkovska T, Salajeghe S, Vanreusel V, Van Audekerke J, Verschuuren M, Hirschler L, Warnking J, Pintelon I, Pustina D, Cachope R, Mrzljak L, Muñoz-Sanjuan I, Barbier EL, De Vos WH, Van der Linden A, Verhoye M. Longitudinal alterations in brain perfusion and vascular reactivity in the zQ175DN mouse model of Huntington's disease. J Biomed Sci 2024; 31:37. [PMID: 38627751 PMCID: PMC11022401 DOI: 10.1186/s12929-024-01028-3] [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: 01/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is marked by a CAG-repeat expansion in the huntingtin gene that causes neuronal dysfunction and loss, affecting mainly the striatum and the cortex. Alterations in the neurovascular coupling system have been shown to lead to dysregulated energy supply to brain regions in several neurological diseases, including HD, which could potentially trigger the process of neurodegeneration. In particular, it has been observed in cross-sectional human HD studies that vascular alterations are associated to impaired cerebral blood flow (CBF). To assess whether whole-brain changes in CBF are present and follow a pattern of progression, we investigated both resting-state brain perfusion and vascular reactivity longitudinally in the zQ175DN mouse model of HD. METHODS Using pseudo-continuous arterial spin labelling (pCASL) MRI in the zQ175DN model of HD and age-matched wild-type (WT) mice, we assessed whole-brain, resting-state perfusion at 3, 6 and 9 and 13 months of age, and assessed hypercapnia-induced cerebrovascular reactivity (CVR), at 4.5, 6, 9 and 15 months of age. RESULTS We found increased perfusion in cortical regions of zQ175DN HET mice at 3 months of age, and a reduction of this anomaly at 6 and 9 months, ages at which behavioural deficits have been reported. On the other hand, under hypercapnia, CBF was reduced in zQ175DN HET mice as compared to the WT: for multiple brain regions at 6 months of age, for only somatosensory and retrosplenial cortices at 9 months of age, and brain-wide by 15 months. CVR impairments in cortical regions, the thalamus and globus pallidus were observed in zQ175DN HET mice at 9 months, with whole brain reactivity diminished at 15 months of age. Interestingly, blood vessel density was increased in the motor cortex at 3 months, while average vessel length was reduced in the lateral portion of the caudate putamen at 6 months of age. CONCLUSION Our findings reveal early cortical resting-state hyperperfusion and impaired CVR at ages that present motor anomalies in this HD model, suggesting that further characterization of brain perfusion alterations in animal models is warranted as a potential therapeutic target in HD.
Collapse
Affiliation(s)
- Tamara Vasilkovska
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium.
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium.
| | - Somaie Salajeghe
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Verdi Vanreusel
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Johan Van Audekerke
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marlies Verschuuren
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Lydiane Hirschler
- C.J. Gorter MRI Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Warnking
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Isabel Pintelon
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Dorian Pustina
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
| | - Roger Cachope
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
| | - Ladislav Mrzljak
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
- Present Address: Takeda Pharmaceuticals, Cambridge, MA, USA
| | - Ignacio Muñoz-Sanjuan
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
- Present Address: Cajal Neuroscience Inc, Seattle, WA, USA
| | - Emmanuel L Barbier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Winnok H De Vos
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Annemie Van der Linden
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
6
|
Gong Y, Laheji F, Berenson A, Li Y, Moser A, Qian A, Frosch M, Sadjadi R, Hahn R, Maguire CA, Eichler F. Role of Basal Forebrain Neurons in Adrenomyeloneuropathy in Mice and Humans. Ann Neurol 2024; 95:442-458. [PMID: 38062617 PMCID: PMC10949091 DOI: 10.1002/ana.26849] [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: 06/05/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 12/27/2023]
Abstract
OBJECTIVE X-linked adrenoleukodystrophy is caused by mutations in the peroxisomal half-transporter ABCD1. The most common manifestation is adrenomyeloneuropathy, a hereditary spastic paraplegia of adulthood. The present study set out to understand the role of neuronal ABCD1 in mice and humans with adrenomyeloneuropathy. METHODS Neuronal expression of ABCD1 during development was assessed in mice and humans. ABCD1-deficient mice and human brain tissues were examined for corresponding pathology. Next, we silenced ABCD1 in cholinergic Sh-sy5y neurons to investigate its impact on neuronal function. Finally, we tested adeno-associated virus vector-mediated ABCD1 delivery to the brain in mice with adrenomyeloneuropathy. RESULTS ABCD1 is highly expressed in neurons located in the periaqueductal gray matter, basal forebrain and hypothalamus. In ABCD1-deficient mice (Abcd1-/y), these structures showed mild accumulations of α-synuclein. Similarly, healthy human controls had high expression of ABCD1 in deep gray nuclei, whereas X-ALD patients showed increased levels of phosphorylated tau, gliosis, and complement activation in those same regions, albeit not to the degree seen in neurodegenerative tauopathies. Silencing ABCD1 in Sh-sy5y neurons impaired expression of functional proteins and decreased acetylcholine levels, similar to observations in plasma of Abcd1-/y mice. Notably, hind limb clasping in Abcd1-/y mice was corrected through transduction of ABCD1 in basal forebrain neurons following intracerebroventricular gene delivery. INTERPRETATION Our study suggests that the basal forebrain-cortical cholinergic pathway may contribute to dysfunction in adrenomyeloneuropathy. Rescuing peroxisomal transport activity in basal forebrain neurons and supporting glial cells might represent a viable therapeutic strategy. ANN NEUROL 2024;95:442-458.
Collapse
Affiliation(s)
- Yi Gong
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Fiza Laheji
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Anna Berenson
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Yedda Li
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Ann Moser
- Peroxisome Disease Lab, Hugo W Moser Research Institute, Baltimore, MD, USA
| | - April Qian
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Matthew Frosch
- Massachusetts General Hospital, Department of Neuropathology, Harvard Medical School, Boston
| | - Reza Sadjadi
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Ryan Hahn
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Casey A. Maguire
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Florian Eichler
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| |
Collapse
|
7
|
Stoklund Dittlau K, Freude K. Astrocytes: The Stars in Neurodegeneration? Biomolecules 2024; 14:289. [PMID: 38540709 PMCID: PMC10967965 DOI: 10.3390/biom14030289] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 11/11/2024] Open
Abstract
Today, neurodegenerative disorders like Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) affect millions of people worldwide, and as the average human lifespan increases, similarly grows the number of patients. For many decades, cognitive and motoric decline has been explained by the very apparent deterioration of neurons in various regions of the brain and spinal cord. However, more recent studies show that disease progression is greatly influenced by the vast population of glial cells. Astrocytes are traditionally considered star-shaped cells on which neurons rely heavily for their optimal homeostasis and survival. Increasing amounts of evidence depict how astrocytes lose their supportive functions while simultaneously gaining toxic properties during neurodegeneration. Many of these changes are similar across various neurodegenerative diseases, and in this review, we highlight these commonalities. We discuss how astrocyte dysfunction drives neuronal demise across a wide range of neurodegenerative diseases, but rather than categorizing based on disease, we aim to provide an overview based on currently known mechanisms. As such, this review delivers a different perspective on the disease causes of neurodegeneration in the hope to encourage further cross-disease studies into shared disease mechanisms, which might ultimately disclose potentially common therapeutic entry points across a wide panel of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark;
| |
Collapse
|
8
|
Pérot JB, Brouillet E, Flament J. The contribution of preclinical magnetic resonance imaging and spectroscopy to Huntington's disease. Front Aging Neurosci 2024; 16:1306312. [PMID: 38414634 PMCID: PMC10896846 DOI: 10.3389/fnagi.2024.1306312] [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: 10/03/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024] Open
Abstract
Huntington's disease is an inherited disorder characterized by psychiatric, cognitive, and motor symptoms due to degeneration of medium spiny neurons in the striatum. A prodromal phase precedes the onset, lasting decades. Current biomarkers include clinical score and striatal atrophy using Magnetic Resonance Imaging (MRI). These markers lack sensitivity for subtle cellular changes during the prodromal phase. MRI and MR spectroscopy offer different contrasts for assessing metabolic, microstructural, functional, or vascular alterations in the disease. They have been used in patients and mouse models. Mouse models can be of great interest to study a specific mechanism of the degenerative process, allow better understanding of the pathogenesis from the prodromal to the symptomatic phase, and to evaluate therapeutic efficacy. Mouse models can be divided into three different constructions: transgenic mice expressing exon-1 of human huntingtin (HTT), mice with an artificial chromosome expressing full-length human HTT, and knock-in mouse models with CAG expansion inserted in the murine htt gene. Several studies have used MRI/S to characterized these models. However, the multiplicity of modalities and mouse models available complicates the understanding of this rich corpus. The present review aims at giving an overview of results obtained using MRI/S for each mouse model of HD, to provide a useful resource for the conception of neuroimaging studies using mouse models of HD. Finally, despite difficulties in translating preclinical protocols to clinical applications, many biomarkers identified in preclinical models have already been evaluated in patients. This review also aims to cover this aspect to demonstrate the importance of MRI/S for studying HD.
Collapse
Affiliation(s)
- Jean-Baptiste Pérot
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
- Institut du Cerveau – Paris Brain Institute – ICM, Sorbonne Université, Paris, France
| | - Emmanuel Brouillet
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Flament
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
9
|
Sharif A, Mamo J, Lam V, Al-Salami H, Mooranian A, Watts GF, Clarnette R, Luna G, Takechi R. The therapeutic potential of probucol and probucol analogues in neurodegenerative diseases. Transl Neurodegener 2024; 13:6. [PMID: 38247000 PMCID: PMC10802046 DOI: 10.1186/s40035-024-00398-w] [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: 11/05/2023] [Accepted: 01/07/2024] [Indexed: 01/23/2024] Open
Abstract
Neurodegenerative disorders present complex pathologies characterized by various interconnected factors, including the aggregation of misfolded proteins, oxidative stress, neuroinflammation and compromised blood-brain barrier (BBB) integrity. Addressing such multifaceted pathways necessitates the development of multi-target therapeutic strategies. Emerging research indicates that probucol, a historic lipid-lowering medication, offers substantial potential in the realm of neurodegenerative disease prevention and treatment. Preclinical investigations have unveiled multifaceted cellular effects of probucol, showcasing its remarkable antioxidative and anti-inflammatory properties, its ability to fortify the BBB and its direct influence on neural preservation and adaptability. These diverse effects collectively translate into enhancements in both motor and cognitive functions. This review provides a comprehensive overview of recent findings highlighting the efficacy of probucol and probucol-related compounds in the context of various neurodegenerative conditions, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Arazu Sharif
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - John Mamo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Research, Perth, WA, Australia
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Hani Al-Salami
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Armin Mooranian
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| | - Roger Clarnette
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| | - Giuseppe Luna
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Ryu Takechi
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.
| |
Collapse
|
10
|
Hernandez SJ, Lim RG, Onur T, Dane MA, Smith R, Wang K, Jean GEH, Reyes-Ortiz A, Devlin K, Miramontes R, Wu J, Casale M, Kilburn D, Heiser LM, Korkola JE, Van Vactor D, Botas J, Thompson-Peer KL, Thompson LM. An altered extracellular matrix-integrin interface contributes to Huntington's disease-associated CNS dysfunction in glial and vascular cells. Hum Mol Genet 2023; 32:1483-1496. [PMID: 36547263 PMCID: PMC10117161 DOI: 10.1093/hmg/ddac303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Astrocytes and brain endothelial cells are components of the neurovascular unit that comprises the blood-brain barrier (BBB) and their dysfunction contributes to pathogenesis in Huntington's disease (HD). Defining the contribution of these cells to disease can inform cell-type-specific effects and uncover new disease-modifying therapeutic targets. These cells express integrin (ITG) adhesion receptors that anchor the cells to the extracellular matrix (ECM) to maintain the integrity of the BBB. We used HD patient-derived induced pluripotent stem cell (iPSC) modeling to study the ECM-ITG interface in astrocytes and brain microvascular endothelial cells and found ECM-ITG dysregulation in human iPSC-derived cells that may contribute to the dysfunction of the BBB in HD. This disruption has functional consequences since reducing ITG expression in glia in an HD Drosophila model suppressed disease-associated CNS dysfunction. Since ITGs can be targeted therapeutically and manipulating ITG signaling prevents neurodegeneration in other diseases, defining the role of ITGs in HD may provide a novel strategy of intervention to slow CNS pathophysiology to treat HD.
Collapse
Affiliation(s)
- Sarah J Hernandez
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Ryan G Lim
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Tarik Onur
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mark A Dane
- Department of Biomedical Engineering, OHSU, Portland, OR 97239, USA
| | - Rebecca Smith
- Department of Biomedical Engineering, OHSU, Portland, OR 97239, USA
| | - Keona Wang
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Grace En-Hway Jean
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Andrea Reyes-Ortiz
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Kaylyn Devlin
- Department of Biomedical Engineering, OHSU, Portland, OR 97239, USA
| | - Ricardo Miramontes
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Malcolm Casale
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - David Kilburn
- Department of Biomedical Engineering, OHSU, Portland, OR 97239, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, OHSU, Portland, OR 97239, USA
- OHSU Knight Cancer Institute, Portland, OR 97239, USA
| | - James E Korkola
- Department of Biomedical Engineering, OHSU, Portland, OR 97239, USA
- OHSU Knight Cancer Institute, Portland, OR 97239, USA
| | - David Van Vactor
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Katherine L Thompson-Peer
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Reeve-Irvine Research Center, University of California, Irvine, CA 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Reyes-Ortiz AM, Abud EM, Burns MS, Wu J, Hernandez SJ, McClure N, Wang KQ, Schulz CJ, Miramontes R, Lau A, Michael N, Miyoshi E, Van Vactor D, Reidling JC, Blurton-Jones M, Swarup V, Poon WW, Lim RG, Thompson LM. Single-nuclei transcriptome analysis of Huntington disease iPSC and mouse astrocytes implicates maturation and functional deficits. iScience 2023; 26:105732. [PMID: 36590162 PMCID: PMC9800269 DOI: 10.1016/j.isci.2022.105732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/13/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by expanded CAG repeats in the huntingtin gene that alters cellular homeostasis, particularly in the striatum and cortex. Astrocyte signaling that establishes and maintains neuronal functions are often altered under pathological conditions. We performed single-nuclei RNA-sequencing on human HD patient-induced pluripotent stem cell (iPSC)-derived astrocytes and on striatal and cortical tissue from R6/2 HD mice to investigate high-resolution HD astrocyte cell state transitions. We observed altered maturation and glutamate signaling in HD human and mouse astrocytes. Human HD astrocytes also showed upregulated actin-mediated signaling, suggesting that some states may be cell-autonomous and human specific. In both species, astrogliogenesis transcription factors may drive HD astrocyte maturation deficits, which are supported by rescued climbing deficits in HD drosophila with NFIA knockdown. Thus, dysregulated HD astrocyte states may induce dysfunctional astrocytic properties, in part due to maturation deficits influenced by astrogliogenesis transcription factor dysregulation.
Collapse
Affiliation(s)
- Andrea M. Reyes-Ortiz
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92617, USA
| | - Edsel M. Abud
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - Mara S. Burns
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92617, USA
| | - Sarah J. Hernandez
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - Nicolette McClure
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92617, USA
| | - Keona Q. Wang
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - Corey J. Schulz
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92617, USA
| | - Ricardo Miramontes
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| | - Alice Lau
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - Neethu Michael
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - Emily Miyoshi
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
| | - David Van Vactor
- Harvard Medical School, Department of Cell Biology, Boston, MA 02115, USA
| | - John C. Reidling
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92617, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| | - Vivek Swarup
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| | - Wayne W. Poon
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| | - Ryan G. Lim
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| | - Leslie M. Thompson
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92617, USA
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92617, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92617, USA
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA 92617, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92617, USA
| |
Collapse
|
12
|
Ishihara K, Takata K, Mizutani KI. Involvement of an Aberrant Vascular System in Neurodevelopmental, Neuropsychiatric, and Neuro-Degenerative Diseases. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010221. [PMID: 36676170 PMCID: PMC9866034 DOI: 10.3390/life13010221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The vascular system of the prenatal brain is crucial for the development of the central nervous system. Communication between vessels and neural cells is bidirectional, and dysfunctional communication can lead to neurodevelopmental diseases. In the present review, we introduce neurodevelopmental and neuropsychiatric diseases potentially caused by disturbances in the neurovascular system and discuss candidate genes responsible for neurovascular system impairments. In contrast to diseases that can manifest during the developing stage, we have also summarized the disturbances of the neurovascular system in neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. Furthermore, we discussed the role of abnormal vascularization and dysfunctional vessels in the development of neurovascular-related diseases.
Collapse
Affiliation(s)
- Keiichi Ishihara
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
- Correspondence: ; Tel.: +81-75-595-4656
| | - Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Ken-ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| |
Collapse
|
13
|
Abjean L, Ben Haim L, Riquelme-Perez M, Gipchtein P, Derbois C, Palomares MA, Petit F, Hérard AS, Gaillard MC, Guillermier M, Gaudin-Guérif M, Aurégan G, Sagar N, Héry C, Dufour N, Robil N, Kabani M, Melki R, De la Grange P, Bemelmans AP, Bonvento G, Deleuze JF, Hantraye P, Flament J, Bonnet E, Brohard S, Olaso R, Brouillet E, Carrillo-de Sauvage MA, Escartin C. Reactive astrocytes promote proteostasis in Huntington's disease through the JAK2-STAT3 pathway. Brain 2023; 146:149-166. [PMID: 35298632 DOI: 10.1093/brain/awac068] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 01/11/2023] Open
Abstract
Huntington's disease is a fatal neurodegenerative disease characterized by striatal neurodegeneration, aggregation of mutant Huntingtin and the presence of reactive astrocytes. Astrocytes are important partners for neurons and engage in a specific reactive response in Huntington's disease that involves morphological, molecular and functional changes. How reactive astrocytes contribute to Huntington's disease is still an open question, especially because their reactive state is poorly reproduced in experimental mouse models. Here, we show that the JAK2-STAT3 pathway, a central cascade controlling astrocyte reactive response, is activated in the putamen of Huntington's disease patients. Selective activation of this cascade in astrocytes through viral gene transfer reduces the number and size of mutant Huntingtin aggregates in neurons and improves neuronal defects in two complementary mouse models of Huntington's disease. It also reduces striatal atrophy and increases glutamate levels, two central clinical outcomes measured by non-invasive magnetic resonance imaging. Moreover, astrocyte-specific transcriptomic analysis shows that activation of the JAK2-STAT3 pathway in astrocytes coordinates a transcriptional program that increases their intrinsic proteolytic capacity, through the lysosomal and ubiquitin-proteasome degradation systems. This pathway also enhances their production and exosomal release of the co-chaperone DNAJB1, which contributes to mutant Huntingtin clearance in neurons. Together, our results show that the JAK2-STAT3 pathway controls a beneficial proteostasis response in reactive astrocytes in Huntington's disease, which involves bi-directional signalling with neurons to reduce mutant Huntingtin aggregation, eventually improving disease outcomes.
Collapse
Affiliation(s)
- Laurene Abjean
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Lucile Ben Haim
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Miriam Riquelme-Perez
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France.,Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Pauline Gipchtein
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Céline Derbois
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Marie-Ange Palomares
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Fanny Petit
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Martine Guillermier
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Mylène Gaudin-Guérif
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Gwennaëlle Aurégan
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Nisrine Sagar
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Cameron Héry
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Noëlle Dufour
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | | | - Mehdi Kabani
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Ronald Melki
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | | | - Alexis P Bemelmans
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Gilles Bonvento
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Jean-François Deleuze
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Philippe Hantraye
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Julien Flament
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Eric Bonnet
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Solène Brohard
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Robert Olaso
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Emmanuel Brouillet
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Maria-Angeles Carrillo-de Sauvage
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| | - Carole Escartin
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265 Fontenay-aux-Roses, France
| |
Collapse
|
14
|
Yuan Y, Sun J, Dong Q, Cui M. Blood-brain barrier endothelial cells in neurodegenerative diseases: Signals from the "barrier". Front Neurosci 2023; 17:1047778. [PMID: 36908787 PMCID: PMC9998532 DOI: 10.3389/fnins.2023.1047778] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
As blood-brain barrier (BBB) disruption emerges as a common problem in the early stages of neurodegenerative diseases, the crucial roles of barrier-type brain endothelial cells (BECs), the primary part of the BBB, have been reported in the pathophysiology of neurodegenerative diseases. The mechanisms of how early vascular dysfunction contributes to the progress of neurodegeneration are still unclear, and understanding BEC functions is a promising start. Our understanding of the BBB has gone through different stages, from a passive diffusion barrier to a mediator of central-peripheral interactions. BECs serve two seemingly paradoxical roles: as a barrier to protect the delicate brain from toxins and as an interface to constantly receive and release signals, thus maintaining and regulating the homeostasis of the brain. Most previous studies about neurodegenerative diseases focus on the loss of barrier functions, and far too little attention has been paid to the active regulations of BECs. In this review, we present the current evidence of BEC dysfunction in neurodegenerative diseases and explore how BEC signals participate in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiwen Yuan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Sun
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Modelling the Human Blood-Brain Barrier in Huntington Disease. Int J Mol Sci 2022; 23:ijms23147813. [PMID: 35887162 PMCID: PMC9321930 DOI: 10.3390/ijms23147813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 02/05/2023] Open
Abstract
While blood–brain barrier (BBB) dysfunction has been described in neurological disorders, including Huntington’s disease (HD), it is not known if endothelial cells themselves are functionally compromised when promoting BBB dysfunction. Furthermore, the underlying mechanisms of BBB dysfunction remain elusive given the limitations with mouse models and post mortem tissue to identify primary deficits. We established models of BBB and undertook a transcriptome and functional analysis of human induced pluripotent stem cell (iPSC)-derived brain-like microvascular endothelial cells (iBMEC) from HD patients or unaffected controls. We demonstrated that HD-iBMECs have abnormalities in barrier properties, as well as in specific BBB functions such as receptor-mediated transcytosis.
Collapse
|
16
|
Linville RM, Nerenberg RF, Grifno G, Arevalo D, Guo Z, Searson PC. Brain microvascular endothelial cell dysfunction in an isogenic juvenile iPSC model of Huntington's disease. Fluids Barriers CNS 2022; 19:54. [PMID: 35773691 PMCID: PMC9245306 DOI: 10.1186/s12987-022-00347-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease caused by expansion of cytosine-adenine-guanine (CAG) repeats in the huntingtin gene, which leads to neuronal loss and decline in cognitive and motor function. Increasing evidence suggests that blood-brain barrier (BBB) dysfunction may contribute to progression of the disease. Studies in animal models, in vitro models, and post-mortem tissue find that disease progression is associated with increased microvascular density, altered cerebral blood flow, and loss of paracellular and transcellular barrier function. Here, we report on changes in BBB phenotype due to expansion of CAG repeats using an isogenic pair of induced pluripotent stem cells (iPSCs) differentiated into brain microvascular endothelial-like cells (iBMECs). We show that CAG expansion associated with juvenile HD alters the trajectory of iBMEC differentiation, producing cells with ~ two-fold lower percentage of adherent endothelial cells. CAG expansion is associated with diminished transendothelial electrical resistance and reduced tight junction protein expression, but no significant changes in paracellular permeability. While mutant huntingtin protein (mHTT) aggregates were not observed in HD iBMECs, widespread transcriptional dysregulation was observed in iBMECs compared to iPSCs. In addition, CAG expansion in iBMECs results in distinct responses to pathological and therapeutic perturbations including angiogenic factors, oxidative stress, and osmotic stress. In a tissue-engineered BBB model, iBMECs show subtle changes in phenotype, including differences in cell turnover and immune cell adhesion. Our results further support that CAG expansion in BMECs contributes to BBB dysfunction during HD.
Collapse
Affiliation(s)
- Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Renée F Nerenberg
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielle Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Diego Arevalo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
17
|
Zhang J, Liu Z, Wu H, Chen X, Hu Q, Li X, Luo L, Ye S, Ye J. Irisin Attenuates Pathological Neovascularization in Oxygen-Induced Retinopathy Mice. Invest Ophthalmol Vis Sci 2022; 63:21. [PMID: 35737379 PMCID: PMC9233294 DOI: 10.1167/iovs.63.6.21] [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] [Indexed: 11/25/2022] Open
Abstract
Purpose Abnormal angiogenesis is a defining feature in a couple of ocular neovascular diseases. The application of anti-VEGFA therapy has achieved certain benefits in the clinic, accompanying side effects and poor responsiveness in many patients. The present study investigated the role of irisin in retinal neovascularization. Methods Western blot and quantitative PCR were used to determine irisin expression in the oxygen-induced retinopathy mice model. The pathological angiogenesis and inflammation index were examined after irisin administration. Primary retinal astrocytes were cultured and analyzed for VEGFA expression in vitro. Astrocyte-conditioned medium was collected for transwell assay and tube formation assay in human microvascular endothelial cells-1. Results Irisin was downregulated in the oxygen-induced retinopathy mice retinae. Additional irisin attenuated pathological angiogenesis, inflammation, and apoptosis in vivo. In vitro, irisin decreased astrocyte VEGFA production, and the conditioned medium suppressed human microvascular endothelial cells-1 migration. Last, irisin inhibited hypoxia-inducible factor-2α, nuclear factor-κB, and pNF-κB (Phospho-Nuclear Factor-κB) expression. Conclusions Irisin mitigates retinal pathological angiogenesis. Chinese Abstract
目的:异常的血管生成是新生血管性眼病的显著特征。抗血管内皮生长因子A的治疗在临床上取得了一定的效果, 然而同时伴随着不可避免的副作用和不良反应。本研究旨在探讨irisin在视网膜病理性新生血管形成中的作用。
方法:采用免疫印迹和qPCR检测氧诱导视网膜病变小鼠模型中irisin的表达。外源性给予irisin后, 检测病理性血管生成和炎症的相关指标。为了研究irisin在体外的作用, 我们培养了原代视网膜星形胶质细胞, 检测缺氧后VEGFA的表达, 并收集星形胶质细胞的条件培养基用于人微血管内皮细胞-1(HMEC-1)的迁移和管腔形成实验。
结果:irisin在氧诱导视网膜病变小鼠视网膜中下调。外源性加入irisin可抑制病理性血管生成、炎症和凋亡。在体外, irisin减少星形胶质细胞中VEGFA的生成, 其处理过的星形胶质细胞条件培养基可以抑制人微血管内皮细胞-1的迁移。最后, 我们发现irisin可以降低HIF-2α、NF-κB和pNF-κB的表达水平。
结论:irisin可减轻视网膜病理性血管生成。
Collapse
Affiliation(s)
- Jieqiong Zhang
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Zhifei Liu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Haoqian Wu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Qiumei Hu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xue Li
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Linlin Luo
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Shiyang Ye
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
18
|
Martínez-Lazcano JC, González-Guevara E, Boll C, Cárdenas G. Gut dysbiosis and homocysteine: a couple for boosting neurotoxicity in Huntington disease. Rev Neurosci 2022; 33:819-827. [PMID: 35411760 DOI: 10.1515/revneuro-2021-0164] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/11/2022] [Indexed: 11/15/2022]
Abstract
Huntington's disease (HD), a neurodegenerative disorder caused by an expansion of the huntingtin triplet (Htt), is clinically characterized by cognitive and neuropsychiatric alterations. Although these alterations appear to be related to mutant Htt (mHtt)-induced neurotoxicity, several other factors are involved. The gut microbiota is a known modulator of brain-gut communication and when altered (dysbiosis), several complaints can be developed including gastrointestinal dysfunction which may have a negative impact on cognition, behavior, and other mental functions in HD through several mechanisms, including increased levels of lipopolysaccharide, proinflammatory cytokines and immune cell response, as well as alterations in Ca2+ signaling, resulting in both increased intestinal and blood-brain barrier (BBB) permeability. Recently, the presence of dysbiosis has been described in both transgenic mouse models and HD patients. A bidirectional influence between host brain tissues and the gut microbiota has been observed. On the one hand, the host diet influences the composition and function of microbiota; and on the other hand, microbiota products can affect BBB permeability, synaptogenesis, and the regulation of neurotransmitters and neurotrophic factors, which has a direct effect on host metabolism and brain function. This review summarizes the available evidence on the pathogenic synergism of dysbiosis and homocysteine, and their role in the transgression of BBB integrity and their potential neurotoxicity of HD.
Collapse
Affiliation(s)
- Juan Carlos Martínez-Lazcano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía MVS, Mexico City 14629, Mexico
| | - Edith González-Guevara
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía MVS, Mexico City 14629, Mexico
| | - Catherine Boll
- Laboratorio de Investigación clínica, Clínica de Ataxias y Coreas, Enfermedades Neurodegenerativas Raras, Instituto Nacional de Neurología y Neurocirugía MVS, Mexico City 14629, Mexico
| | - Graciela Cárdenas
- Departamento de Neurología y Enfermedades Neuro-Infecciosas, Instituto Nacional de Neurología y Neurocirugía MVS, Mexico City 14629, Mexico
| |
Collapse
|
19
|
The blood-brain barrier in aging and neurodegeneration. Mol Psychiatry 2022; 27:2659-2673. [PMID: 35361905 PMCID: PMC9156404 DOI: 10.1038/s41380-022-01511-z] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/24/2022] [Accepted: 02/24/2022] [Indexed: 12/01/2022]
Abstract
The blood-brain barrier (BBB) is vital for maintaining brain homeostasis by enabling an exquisite control of exchange of compounds between the blood and the brain parenchyma. Moreover, the BBB prevents unwanted toxins and pathogens from entering the brain. This barrier, however, breaks down with age and further disruption is a hallmark of many age-related disorders. Several drugs have been explored, thus far, to protect or restore BBB function. With the recent connection between the BBB and gut microbiota, microbial-derived metabolites have been explored for their capabilities to protect and restore BBB physiology. This review, will focus on the vital components that make up the BBB, dissect levels of disruption of the barrier, and discuss current drugs and therapeutics that maintain barrier integrity and the recent discoveries of effects microbial-derived metabolites have on BBB physiology.
Collapse
|
20
|
Zisis E, Keller D, Kanari L, Arnaudon A, Gevaert M, Delemontex T, Coste B, Foni A, Abdellah M, Calì C, Hess K, Magistretti PJ, Schürmann F, Markram H. Digital Reconstruction of the Neuro-Glia-Vascular Architecture. Cereb Cortex 2021; 31:5686-5703. [PMID: 34387659 PMCID: PMC8568010 DOI: 10.1093/cercor/bhab254] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 01/21/2023] Open
Abstract
Astrocytes connect the vasculature to neurons mediating the supply of nutrients and biochemicals. They are involved in a growing number of physiological and pathophysiological processes that result from biophysical, physiological, and molecular interactions in this neuro-glia-vascular ensemble (NGV). The lack of a detailed cytoarchitecture severely restricts the understanding of how they support brain function. To address this problem, we used data from multiple sources to create a data-driven digital reconstruction of the NGV at micrometer anatomical resolution. We reconstructed 0.2 mm3 of the rat somatosensory cortex with 16 000 morphologically detailed neurons, 2500 protoplasmic astrocytes, and its microvasculature. The consistency of the reconstruction with a wide array of experimental measurements allows novel predictions of the NGV organization, allowing the anatomical reconstruction of overlapping astrocytic microdomains and the quantification of endfeet connecting each astrocyte to the vasculature, as well as the extent to which they cover the latter. Structural analysis showed that astrocytes optimize their positions to provide uniform vascular coverage for trophic support and signaling. However, this optimal organization rapidly declines as their density increases. The NGV digital reconstruction is a resource that will enable a better understanding of the anatomical principles and geometric constraints, which govern how astrocytes support brain function.
Collapse
Affiliation(s)
- Eleftherios Zisis
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Daniel Keller
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Lida Kanari
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Alexis Arnaudon
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Michael Gevaert
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Thomas Delemontex
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Benoît Coste
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Alessandro Foni
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Marwan Abdellah
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Corrado Calì
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Turin 10043, Italy
- Department of Neuroscience, University of Torino, Torino 10126, Italy
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Kathryn Hess
- Laboratory for Topology and Neuroscience, Brain Mind Institute, École polytechnique fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Pierre Julius Magistretti
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Felix Schürmann
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, Geneva 1202, Switzerland
| |
Collapse
|
21
|
Ouellette J, Lacoste B. From Neurodevelopmental to Neurodegenerative Disorders: The Vascular Continuum. Front Aging Neurosci 2021; 13:749026. [PMID: 34744690 PMCID: PMC8570842 DOI: 10.3389/fnagi.2021.749026] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Structural and functional integrity of the cerebral vasculature ensures proper brain development and function, as well as healthy aging. The inability of the brain to store energy makes it exceptionally dependent on an adequate supply of oxygen and nutrients from the blood stream for matching colossal demands of neural and glial cells. Key vascular features including a dense vasculature, a tightly controlled environment, and the regulation of cerebral blood flow (CBF) all take part in brain health throughout life. As such, healthy brain development and aging are both ensured by the anatomical and functional interaction between the vascular and nervous systems that are established during brain development and maintained throughout the lifespan. During critical periods of brain development, vascular networks remodel until they can actively respond to increases in neural activity through neurovascular coupling, which makes the brain particularly vulnerable to neurovascular alterations. The brain vasculature has been strongly associated with the onset and/or progression of conditions associated with aging, and more recently with neurodevelopmental disorders. Our understanding of cerebrovascular contributions to neurological disorders is rapidly evolving, and increasing evidence shows that deficits in angiogenesis, CBF and the blood-brain barrier (BBB) are causally linked to cognitive impairment. Moreover, it is of utmost curiosity that although neurodevelopmental and neurodegenerative disorders express different clinical features at different stages of life, they share similar vascular abnormalities. In this review, we present an overview of vascular dysfunctions associated with neurodevelopmental (autism spectrum disorders, schizophrenia, Down Syndrome) and neurodegenerative (multiple sclerosis, Huntington's, Parkinson's, and Alzheimer's diseases) disorders, with a focus on impairments in angiogenesis, CBF and the BBB. Finally, we discuss the impact of early vascular impairments on the expression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Julie Ouellette
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
22
|
Chan ST, Mercaldo ND, Kwong KK, Hersch SM, Rosas HD. Impaired Cerebrovascular Reactivity in Huntington's Disease. Front Physiol 2021; 12:663898. [PMID: 34366879 PMCID: PMC8334185 DOI: 10.3389/fphys.2021.663898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
There is increasing evidence that impairments of cerebrovascular function and/or abnormalities of the cerebral vasculature might contribute to early neuronal cell loss in Huntington's disease (HD). Studies in both healthy individuals as well as in patients with other neurodegenerative disorders have used an exogenous carbon dioxide (CO2) challenge in conjunction with functional magnetic resonance imaging (fMRI) to assess regional cerebrovascular reactivity (CVR). In this study, we explored potential impairments of CVR in HD. Twelve gene expanded HD individuals, including both pre-symptomatic and early symptomatic HD and eleven healthy controls were administered a gas mixture targeting a 4-8 mmHg increase in CO2 relative to the end-tidal partial pressure of CO2 (P ET CO2) at rest. A Hilbert Transform analysis was used to compute the cross-correlation between the time series of regional BOLD signal changes (ΔBOLD) and increased P ET CO2, and to estimate the response delay of ΔBOLD relative to P ET CO2. After correcting for age, we found that the cross-correlation between the time series for regional ΔBOLD and for P ET CO2 was weaker in HD subjects than in controls in several subcortical white matter regions, including the corpus callosum, subcortical white matter adjacent to rostral and caudal anterior cingulate, rostral and caudal middle frontal, insular, middle temporal, and posterior cingulate areas. In addition, greater volume of dilated perivascular space (PVS) was observed to overlap, primarily along the periphery, with the areas that showed greater ΔBOLD response delay. Our preliminary findings support that alterations in cerebrovascular function occur in HD and may be an important, not as yet considered, contributor to early neuropathology in HD.
Collapse
Affiliation(s)
- Suk Tak Chan
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Nathaniel D Mercaldo
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Kenneth K Kwong
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Steven M Hersch
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Herminia D Rosas
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Wu YC, Sonninen TM, Peltonen S, Koistinaho J, Lehtonen Š. Blood-Brain Barrier and Neurodegenerative Diseases-Modeling with iPSC-Derived Brain Cells. Int J Mol Sci 2021; 22:7710. [PMID: 34299328 PMCID: PMC8307585 DOI: 10.3390/ijms22147710] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) regulates the delivery of oxygen and important nutrients to the brain through active and passive transport and prevents neurotoxins from entering the brain. It also has a clearance function and removes carbon dioxide and toxic metabolites from the central nervous system (CNS). Several drugs are unable to cross the BBB and enter the CNS, adding complexity to drug screens targeting brain disorders. A well-functioning BBB is essential for maintaining healthy brain tissue, and a malfunction of the BBB, linked to its permeability, results in toxins and immune cells entering the CNS. This impairment is associated with a variety of neurological diseases, including Alzheimer's disease and Parkinson's disease. Here, we summarize current knowledge about the BBB in neurodegenerative diseases. Furthermore, we focus on recent progress of using human-induced pluripotent stem cell (iPSC)-derived models to study the BBB. We review the potential of novel stem cell-based platforms in modeling the BBB and address advances and key challenges of using stem cell technology in modeling the human BBB. Finally, we highlight future directions in this area.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Tuuli-Maria Sonninen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Sanni Peltonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
24
|
Liu Y, Chen D, Smith A, Ye Q, Gao Y, Zhang W. Three-dimensional remodeling of functional cerebrovascular architecture and gliovascular unit in leptin receptor-deficient mice. J Cereb Blood Flow Metab 2021; 41:1547-1562. [PMID: 33818188 PMCID: PMC8221780 DOI: 10.1177/0271678x211006596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022]
Abstract
The cerebrovascular sequelae of diabetes render victims more susceptible to ischemic stroke, vascular cognitive impairment, and Alzheimer's disease. However, limited knowledge exists on the progressive changes in cerebrovascular structure and functional remodeling in type 2 diabetes. To ascertain the impact of diabetes on whole-brain cerebrovascular perfusion, leptin-receptor-deficient mice were transcardially injected with tomato-lectin before sacrifice. The whole brain was clarified by the Fast free-of-acrylamide clearing tissue technique. Functional vascular anatomy of the cerebrum was visualized by light-sheet microscopy, followed by analysis in Imaris software. We observed enhanced neovascularization in adult db/db mice, characterized by increased branch level and loop structures. Microvascular hypoperfusion was initially detected in juvenile db/db mice, suggesting early onset of insufficient microcirculation. Furthermore, gliovascular unit remodeling was verified by loss of pericytes and overactivation of microglia and astrocytes in adult diabetic mice. However, the integrity of the blood-brain barrier (BBB) was fundamentally preserved, as shown by a lack of extravasation of IgG into the brain parenchyma. In summary, we, for the first time, reveal that functional cerebrovascular remodeling occurs as early as four weeks in db/db mice and the deficit in gliovascular coupling may play a role in cerebral hypoperfusion before BBB breakdown in 16-week-old db/db mice.
Collapse
Affiliation(s)
- Yaan Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Di Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Amanda Smith
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Qing Ye
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wenting Zhang
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Trujillo-Estrada L, Gomez-Arboledas A, Forner S, Martini AC, Gutierrez A, Baglietto-Vargas D, LaFerla FM. Astrocytes: From the Physiology to the Disease. Curr Alzheimer Res 2020; 16:675-698. [PMID: 31470787 DOI: 10.2174/1567205016666190830110152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
Abstract
Astrocytes are key cells for adequate brain formation and regulation of cerebral blood flow as well as for the maintenance of neuronal metabolism, neurotransmitter synthesis and exocytosis, and synaptic transmission. Many of these functions are intrinsically related to neurodegeneration, allowing refocusing on the role of astrocytes in physiological and neurodegenerative states. Indeed, emerging evidence in the field indicates that abnormalities in the astrocytic function are involved in the pathogenesis of multiple neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD) and Amyotrophic Lateral Sclerosis (ALS). In the present review, we highlight the physiological role of astrocytes in the CNS, including their communication with other cells in the brain. Furthermore, we discuss exciting findings and novel experimental approaches that elucidate the role of astrocytes in multiple neurological disorders.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Stefânia Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Antonia Gutierrez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| |
Collapse
|
26
|
Huang JB, Hsu SP, Pan HY, Chen SD, Chen SF, Lin TK, Liu XP, Li JH, Chen NC, Liou CW, Hsu CY, Chuang HY, Chuang YC. Peroxisome Proliferator-Activated Receptor γ Coactivator 1α Activates Vascular Endothelial Growth Factor That Protects Against Neuronal Cell Death Following Status Epilepticus through PI3K/AKT and MEK/ERK Signaling. Int J Mol Sci 2020; 21:ijms21197247. [PMID: 33008083 PMCID: PMC7583914 DOI: 10.3390/ijms21197247] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Status epilepticus may cause molecular and cellular events, leading to hippocampal neuronal cell death. Peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) is an important regulator of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2), also known as fetal liver kinase receptor 1 (Flk-1). Resveratrol is an activator of PGC-1α. It has been suggested to provide neuroprotective effects in epilepsy, stroke, and neurodegenerative diseases. In the present study, we used microinjection of kainic acid into the left hippocampal CA3 region in Sprague Dawley rats to induce bilateral prolonged seizure activity. Upregulating the PGC-1α pathway will increase VEGF/VEGFR2 (Flk-1) signaling and further activate some survival signaling that includes the mitogen activated protein kinase kinase (MEK)/mitogen activated protein kinase (ERK) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways and offer neuroprotection as a consequence of apoptosis in the hippocampal neurons following status epilepticus. Otherwise, downregulation of PGC-1α by siRNA against pgc-1α will inhibit VEGF/VEGFR2 (Flk-1) signaling and suppress pro-survival PI3K/AKT and MEK/ERK pathways that are also accompanied by hippocampal CA3 neuronal cell apoptosis. These results may indicate that the PGC-1α induced VEGF/VEGFR2 pathway may trigger the neuronal survival signaling, and the PI3K/AKT and MEK/ERK signaling pathways. Thus, the axis of PGC-1α/VEGF/VEGFR2 (Flk-1) and the triggering of downstream PI3K/AKT and MEK/ERK signaling could be considered an endogenous neuroprotective effect against apoptosis in the hippocampus following status epilepticus.
Collapse
Affiliation(s)
- Jyun-Bin Huang
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (J.-B.H.); (H.-Y.P.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
| | - Shih-Pin Hsu
- Department of Neurology, E-Da Hospital/School of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Hsiu-Yung Pan
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (J.-B.H.); (H.-Y.P.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
| | - Shang-Der Chen
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
| | - Shu-Fang Chen
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Tsu-Kung Lin
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Xuan-Ping Liu
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
| | - Jie-Hau Li
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
| | - Nai-Ching Chen
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chia-Wei Liou
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chung-Yao Hsu
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Hung-Yi Chuang
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital and School of Public Health, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yao-Chung Chuang
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Correspondence:
| |
Collapse
|
27
|
Rajib D. Central nervous system diseases associated with blood brain barrier breakdown - A Comprehensive update of existing literatures. ACTA ACUST UNITED AC 2020. [DOI: 10.29328/journal.jnnd.1001035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Blood vessels that supply and feed the central nervous system (CNS) possess unique and exclusive properties, named as blood–brain barrier (BBB). It is responsible for tight regulation of the movement of ions, molecules, and cells between the blood and the brain thereby maintaining controlled chemical composition of the neuronal milieu required for appropriate functioning. It also protects the neural tissue from toxic plasma components, blood cells and pathogens from entering the brain. In this review the importance of BBB and its disruption causing brain pathology and progression to different neurological diseases like Alzheimer’s disease (AD), Parkinson’s disease (PD), Amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD) etc. will be discussed.
Collapse
|
28
|
The Role of Neurovascular System in Neurodegenerative Diseases. Mol Neurobiol 2020; 57:4373-4393. [PMID: 32725516 DOI: 10.1007/s12035-020-02023-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
The neurovascular system (NVS), which consisted of neurons, glia, and vascular cells, is a functional and structural unit of the brain. The NVS regulates blood-brain barrier (BBB) permeability and cerebral blood flow (CBF), thereby maintaining the brain's microenvironment for normal functioning, neuronal survival, and information processing. Recent studies have highlighted the role of vascular dysfunction in several neurodegenerative diseases. This is not unexpected since both nervous and vascular systems are functionally interdependent and show close anatomical apposition, as well as similar molecular pathways. However, despite extensive research, the precise mechanism by which neurovascular dysfunction contributes to neurodegeneration remains incomplete. Therefore, understanding the mechanisms of neurovascular dysfunction in disease conditions may allow us to develop potent and effective therapies for prevention and treatment of neurodegenerative diseases. This review article summarizes the current research in the context of neurovascular signaling associated with neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). We also discuss the potential implication of neurovascular factor as a novel therapeutic target and prognostic marker in patients with neurodegenerative conditions. Graphical Abstract.
Collapse
|
29
|
Steventon JJ, Furby H, Ralph J, O'Callaghan P, Rosser AE, Wise RG, Busse M, Murphy K. Altered cerebrovascular response to acute exercise in patients with Huntington's disease. Brain Commun 2020; 2:fcaa044. [PMID: 32566927 PMCID: PMC7293798 DOI: 10.1093/braincomms/fcaa044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 01/20/2023] Open
Abstract
The objective of this study was to determine whether a single session of exercise was sufficient to induce cerebral adaptations in individuals with Huntington’s disease and to explore the time dynamics of any acute cerebrovascular response. In this case–control study, we employed arterial-spin labelling MRI in 19 Huntington’s disease gene-positive participants (32–65 years, 13 males) and 19 controls (29–63 years, 10 males) matched for age, gender, body mass index and self-reported activity levels, to measure global and regional perfusion in response to 20 min of moderate-intensity cycling. Cerebral perfusion was measured at baseline and 15, 40 and 60 min after exercise cessation. Relative to baseline, we found that cerebral perfusion increased in patients with Huntington’s disease yet was unchanged in control participants in the precentral gyrus (P = 0.016), middle frontal gyrus (P = 0.046) and hippocampus (P = 0.048) 40 min after exercise cessation (+15 to +32.5% change in Huntington’s disease participants, −7.7 to 0.8% change in controls). The length of the disease‐causing trinucleotide repeat expansion in the huntingtin gene predicted the change in the precentral gyrus (P = 0.03) and the intensity of the exercise intervention predicted hippocampal perfusion change in Huntington’s disease participants (P < 0.001). In both groups, exercise increased hippocampal blood flow 60 min after exercise cessation (P = 0.039). These findings demonstrate the utility of acute exercise as a clinically sensitive experimental paradigm to modulate the cerebrovasculature. Twenty minutes of aerobic exercise induced transient cerebrovascular adaptations in the hippocampus and cortex selectively in Huntington’s disease participants and likely represents latent neuropathology not evident at rest.
Collapse
Affiliation(s)
- Jessica J Steventon
- Cardiff University Brain Research Imaging Centre, School of Physics and Astronomy, Cardiff University, Cardiff CF24 4HQ, UK.,Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK
| | - Hannah Furby
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - James Ralph
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - Peter O'Callaghan
- Cardiology Department, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Anne E Rosser
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK.,Cardiff Brain Repair Group, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Richard G Wise
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - Monica Busse
- Centre for Trials Research, Cardiff University, Cardiff CF14 4YS, UK
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre, School of Physics and Astronomy, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
30
|
González-Guevara E, Cárdenas G, Pérez-Severiano F, Martínez-Lazcano JC. Dysregulated Brain Cholesterol Metabolism Is Linked to Neuroinflammation in Huntington's Disease. Mov Disord 2020; 35:1113-1127. [PMID: 32410324 DOI: 10.1002/mds.28089] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease is an autosomal-dominant, neurodegenerative disorder caused by a CAG repeat expansion in exon-1 of the huntingtin gene. Alterations in cholesterol metabolism and distribution have been reported in Huntington's disease, including abnormal interactions between mutant huntingtin and sterol regulatory element-binding proteins, decreased levels of apolipoprotein E/cholesterol/low-density lipoprotein receptor complexes, and alterations in the synthesis of ATP-binding cassette transporter A1. Plasma levels of 24S-hydroxycholestrol, a key intermediary in cholesterol metabolism and a possible marker in neurodegenerative diseases, decreased proportionally to the degree of caudate nucleus atrophy. The interaction of mutant huntingtin with sterol regulatory element-binding proteins is of particular interest given that sterol regulatory element-binding proteins play a dual role: They take part in lipid and cholesterol metabolism, but also in the inflammatory response that induces immune cell migration as well as toxic effects, particularly in astrocytes. This work summarizes current evidence on the metabolic and immune implications of sterol regulatory element-binding protein dysregulation in Huntington's disease, highlighting the potential use of drugs that modulate these alterations. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Edith González-Guevara
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "MVS", Mexico City, Mexico
| | - Graciela Cárdenas
- Departamento de Neurología y Enfermedades Neuro-Infecciosas, Instituto Nacional de Neurología y Neurocirugía "MVS", Mexico City, Mexico
| | - Francisca Pérez-Severiano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "MVS", Mexico City, Mexico
| | - Juan Carlos Martínez-Lazcano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía "MVS", Mexico City, Mexico
| |
Collapse
|
31
|
Steventon JJ, Rosser AE, Hart E, Murphy K. Hypertension, Antihypertensive Use and the Delayed-Onset of Huntington's Disease. Mov Disord 2020; 35:937-946. [PMID: 32017180 PMCID: PMC7317197 DOI: 10.1002/mds.27976] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/15/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
Background Hypertension is a modifiable cardiovascular risk factor implicated in neurodegeneration and dementia risk. In Huntington's disease, a monogenic neurodegenerative disease, autonomic and vascular abnormalities have been reported. This study's objective was to examine the relationship between hypertension and disease severity and progression in Huntington's disease. Methods Using longitudinal data from the largest worldwide observational study of Huntington's disease (n = 14,534), we assessed the relationship between hypertension, disease severity, and rate of clinical progression in Huntington's disease mutation carriers. Propensity score matching was used to statistically match normotensive and hypertensive participants for age, sex, body mass index, ethnicity, and CAG length. Results Huntington's disease patients had a lower prevalence of hypertension compared with age‐matched gene‐negative controls. Huntington's disease patients with hypertension had worse cognitive function, a higher depression score, and more marked motor progression over time compared with Huntington's disease patients without hypertension. However, hypertensive patients taking antihypertensive medication had less motor, cognitive, and functional impairment than Huntington's disease patients with untreated hypertension and a later age of clinical onset compared with untreated hypertensive patients and normotensive individuals with Huntington's disease. Conclusions We report the novel finding that hypertension and antihypertensive medication use are associated with altered disease severity, progression, and clinical onset in patients with Huntington's disease. These findings have implications for the management of hypertension in Huntington's disease and suggest that prospective studies of the symptomatic or disease‐modifying potential of antihypertensives in neurodegenerative diseases are warranted. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jessica J Steventon
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and Astronomy, Maindy Road, Cardiff University, Cardiff, Wales, UK
| | - Anne E Rosser
- Neuroscience and Mental Health Research Institute and Brain Research and Intracerebral Neurotherapeutic (BRAIN) unit, School of Medicine, Cardiff University, Cardiff, Wales, UK.,Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Emma Hart
- Bristol Heart Institute (BHI), Clinical Research and Imaging Centre, School of Physiology, Pharmacology and Neuroscience, Bristol University, Bristol, UK
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and Astronomy, Maindy Road, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
32
|
Neural In Vitro Models for Studying Substances Acting on the Central Nervous System. Handb Exp Pharmacol 2020; 265:111-141. [PMID: 32594299 DOI: 10.1007/164_2020_367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Animal models have been greatly contributing to our understanding of physiology, mechanisms of diseases, and toxicity. Yet, their limitations due to, e.g., interspecies variation are reflected in the high number of drug attrition rates, especially in central nervous system (CNS) diseases. Therefore, human-based neural in vitro models for studying safety and efficacy of substances acting on the CNS are needed. Human iPSC-derived cells offer such a platform with the unique advantage of reproducing the "human context" in vitro by preserving the genetic and molecular phenotype of their donors. Guiding the differentiation of hiPSC into cells of the nervous system and combining them in a 2D or 3D format allows to obtain complex models suitable for investigating neurotoxicity or brain-related diseases with patient-derived cells. This chapter will give an overview over stem cell-based human 2D neuronal and mixed neuronal/astrocyte models, in vitro cultures of microglia, as well as CNS disease models and considers new developments in the field, more specifically the use of brain organoids and 3D bioprinted in vitro models for safety and efficacy evaluation.
Collapse
|
33
|
Palpagama TH, Waldvogel HJ, Faull RLM, Kwakowsky A. The Role of Microglia and Astrocytes in Huntington's Disease. Front Mol Neurosci 2019; 12:258. [PMID: 31708741 PMCID: PMC6824292 DOI: 10.3389/fnmol.2019.00258] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 10/10/2019] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease. HD patients present with movement disorders, behavioral and psychiatric symptoms and cognitive decline. This review summarizes the contribution of microglia and astrocytes to HD pathophysiology. Neuroinflammation in the HD brain is characterized by a reactive morphology in these glial cells. Microglia and astrocytes are critical in regulating neuronal activity and maintaining an optimal milieu for neuronal function. Previous studies provide evidence that activated microglia and reactive astrocytes contribute to HD pathology through transcriptional activation of pro-inflammatory genes to perpetuate a chronic inflammatory state. Reactive astrocytes also display functional changes in glutamate and ion homeostasis and energy metabolism. Astrocytic and microglial changes may further contribute to the neuronal death observed with the progression of HD. Importantly, the degree to which these neuroinflammatory changes are detrimental to neurons and contribute to the progression of HD pathology is not well understood. Furthermore, recent observations provide compelling evidence that activated microglia and astrocytes exert a variety of beneficial functions that are essential for limiting tissue damage and preserving neuronal function in the HD brain. Therefore, a better understanding of the neuroinflammatory environment in the brain in HD may lead to the development of targeted and innovative therapeutic opportunities.
Collapse
Affiliation(s)
- Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
Bogorad MI, DeStefano JG, Linville RM, Wong AD, Searson PC. Cerebrovascular plasticity: Processes that lead to changes in the architecture of brain microvessels. J Cereb Blood Flow Metab 2019; 39:1413-1432. [PMID: 31208241 PMCID: PMC6681538 DOI: 10.1177/0271678x19855875] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The metabolic demands of the brain are met by oxygen and glucose, supplied by a complex hierarchical network of microvessels (arterioles, capillaries, and venules). Transient changes in neural activity are accommodated by local dilation of arterioles or capillaries to increase cerebral blood flow and hence nutrient availability. Transport and communication between the circulation and the brain is regulated by the brain microvascular endothelial cells that form the blood-brain barrier. Under homeostatic conditions, there is very little turnover in brain microvascular endothelial cells, and the cerebrovascular architecture is largely static. However, changes in the brain microenvironment, due to environmental factors, disease, or trauma, can result in additive or subtractive changes in cerebrovascular architecture. Additions occur by angiogenesis or vasculogenesis, whereas subtractions occur by vascular pruning, injury, or endothelial cell death. Here we review the various processes that lead to changes in the cerebrovascular architecture, including sustained changes in the brain microenvironment, development and aging, and injury, disease, and repair.
Collapse
Affiliation(s)
- Max I Bogorad
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jackson G DeStefano
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,3 Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew D Wong
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.,3 Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
35
|
Marottoli FM, Priego M, Flores-Barrera E, Pisharody R, Zaldua S, Fan KD, Ekkurthi GK, Brady ST, Morfini GA, Tseng KY, Tai LM. EGF Treatment Improves Motor Behavior and Cortical GABAergic Function in the R6/2 Mouse Model of Huntington's Disease. Mol Neurobiol 2019; 56:7708-7718. [PMID: 31104296 DOI: 10.1007/s12035-019-1634-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/24/2019] [Indexed: 01/13/2023]
Abstract
Recent evidence indicates that disruption of epidermal growth factor (EGF) signaling by mutant huntingtin (polyQ-htt) may contribute to the onset of behavioral deficits observed in Huntington's disease (HD) through a variety of mechanisms, including cerebrovascular dysfunction. Yet, whether EGF signaling modulates the development of HD pathology and the associated behavioral impairments remain unclear. To gain insight on this issue, we used the R6/2 mouse model of HD to assess the impact of chronic EGF treatment on behavior, and cerebrovascular and cortical neuronal functions. We found that bi-weekly treatment with a low dose of EGF (300 µg/kg, i.p.) for 6 weeks was sufficient to effectively improve motor behavior in R6/2 mice and diminish mortality, compared to vehicle-treated littermates. These beneficial effects of EGF treatment were dissociated from changes in cerebrovascular leakiness, a result that was surprising given that EGF ameliorates this deficit in other neurodegenerative diseases. Rather, the beneficial effect of EGF on R6/2 mice behavior was concomitant with a marked amelioration of cortical GABAergic function. As GABAergic transmission in cortical circuits is disrupted in HD, these novel data suggest a potential mechanistic link between deficits in EGF signaling and GABAergic dysfunction in the progression of HD.
Collapse
Affiliation(s)
- Felecia M Marottoli
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mercedes Priego
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Eden Flores-Barrera
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rohan Pisharody
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Steve Zaldua
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kelly D Fan
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Giri K Ekkurthi
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Scott T Brady
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gerardo A Morfini
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
36
|
Kou ZW, Mo JL, Wu KW, Qiu MH, Huang YL, Tao F, Lei Y, Lv LL, Sun FY. Vascular endothelial growth factor increases the function of calcium-impermeable AMPA receptor GluA2 subunit in astrocytes via activation of protein kinase C signaling pathway. Glia 2019; 67:1344-1358. [PMID: 30883902 PMCID: PMC6594043 DOI: 10.1002/glia.23609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 01/23/2019] [Accepted: 02/19/2019] [Indexed: 01/11/2023]
Abstract
Astrocytic calcium signaling plays pivotal roles in the maintenance of neural functions and neurovascular coupling in the brain. Vascular endothelial growth factor (VEGF), an original biological substance of vessels, regulates the movement of calcium and potassium ions across neuronal membrane. In this study, we investigated whether and how VEGF regulates glutamate-induced calcium influx in astrocytes. We used cultured astrocytes combined with living cell imaging to detect the calcium influx induced by glutamate. We found that VEGF quickly inhibited the glutamate/hypoxia-induced calcium influx, which was blocked by an AMPA receptor antagonist CNQX, but not D-AP5 or UBP310, NMDA and kainate receptor antagonist, respectively. VEGF increased phosphorylation of PKCα and AMPA receptor subunit GluA2 in astrocytes, and these effects were diminished by SU1498 or calphostin C, a PKC inhibitor. With the pHluorin assay, we observed that VEGF significantly increased membrane insertion and expression of GluA2, but not GluA1, in astrocytes. Moreover, siRNA-produced knockdown of GluA2 expression in astrocytes reversed the inhibitory effect of VEGF on glutamate-induced calcium influx. Together, our results suggest that VEGF reduces glutamate-induced calcium influx in astrocytes via enhancing PKCα-mediated GluA2 phosphorylation, which in turn promotes the membrane insertion and expression of GluA2 and causes AMPA receptors to switch from calcium-permeable to calcium-impermeable receptors, thereby inhibiting astrocytic calcium influx. The present study reveals that excitatory neurotransmitter glutamate-mediated astrocytic calcium influx can be regulated by vascular biological factor via activation of AMPA receptor GluA2 subunit and uncovers a novel coupling mechanism between astrocytes and endothelial cells within the neurovascular unit.
Collapse
Affiliation(s)
- Zeng-Wei Kou
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Jia-Lin Mo
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Kun-Wei Wu
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Department of System Biology for Medicine, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Mei-Hong Qiu
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Ya-Lin Huang
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Department of System Biology for Medicine, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - Yu Lei
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Ling-Ling Lv
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Feng-Yan Sun
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China.,Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, PR China.,Department of System Biology for Medicine, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, PR China
| |
Collapse
|
37
|
Logan S, Arzua T, Canfield SG, Seminary ER, Sison SL, Ebert AD, Bai X. Studying Human Neurological Disorders Using Induced Pluripotent Stem Cells: From 2D Monolayer to 3D Organoid and Blood Brain Barrier Models. Compr Physiol 2019; 9:565-611. [PMID: 30873582 PMCID: PMC6705133 DOI: 10.1002/cphy.c180025] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurological disorders have emerged as a predominant healthcare concern in recent years due to their severe consequences on quality of life and prevalence throughout the world. Understanding the underlying mechanisms of these diseases and the interactions between different brain cell types is essential for the development of new therapeutics. Induced pluripotent stem cells (iPSCs) are invaluable tools for neurological disease modeling, as they have unlimited self-renewal and differentiation capacity. Mounting evidence shows: (i) various brain cells can be generated from iPSCs in two-dimensional (2D) monolayer cultures; and (ii) further advances in 3D culture systems have led to the differentiation of iPSCs into organoids with multiple brain cell types and specific brain regions. These 3D organoids have gained widespread attention as in vitro tools to recapitulate complex features of the brain, and (iii) complex interactions between iPSC-derived brain cell types can recapitulate physiological and pathological conditions of blood-brain barrier (BBB). As iPSCs can be generated from diverse patient populations, researchers have effectively applied 2D, 3D, and BBB models to recapitulate genetically complex neurological disorders and reveal novel insights into molecular and genetic mechanisms of neurological disorders. In this review, we describe recent progress in the generation of 2D, 3D, and BBB models from iPSCs and further discuss their limitations, advantages, and future ventures. This review also covers the current status of applications of 2D, 3D, and BBB models in drug screening, precision medicine, and modeling a wide range of neurological diseases (e.g., neurodegenerative diseases, neurodevelopmental disorders, brain injury, and neuropsychiatric disorders). © 2019 American Physiological Society. Compr Physiol 9:565-611, 2019.
Collapse
Affiliation(s)
- Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott G. Canfield
- Department of Cellular & Integrative Physiology, IU School of Medicine-Terre Haute, Terre Haute, IN, USA
| | - Emily R. Seminary
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samantha L. Sison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
38
|
Denis HL, Lamontagne-Proulx J, St-Amour I, Mason SL, Rowley JW, Cloutier N, Tremblay MÈ, Vincent AT, Gould PV, Chouinard S, Weyrich AS, Rondina MT, Barker RA, Boilard E, Cicchetti F. Platelet abnormalities in Huntington's disease. J Neurol Neurosurg Psychiatry 2019; 90:272-283. [PMID: 30567722 PMCID: PMC6518476 DOI: 10.1136/jnnp-2018-318854] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/19/2018] [Accepted: 09/24/2018] [Indexed: 11/20/2022]
Abstract
Huntington's disease (HD) is a hereditary disorder that typically manifests in adulthood with a combination of motor, cognitive and psychiatric problems. The pathology is caused by a mutation in the huntingtin gene which results in the production of an abnormal protein, mutant huntingtin (mHtt). This protein is ubiquitously expressed and known to confer toxicity to multiple cell types. We have recently reported that HD brains are also characterised by vascular abnormalities, which include changes in blood vessel density/diameter as well as increased blood-brain barrier (BBB) leakage. OBJECTIVES Seeking to elucidate the origin of these vascular and BBB abnormalities, we studied platelets that are known to play a role in maintaining the integrity of the vasculature and thrombotic pathways linked to this, given they surprisingly contain the highest concentration of mHtt of all blood cells. METHODS We assessed the functional status of platelets by performing ELISA, western blot and RNA sequencing in a cohort of 71 patients and 68 age- and sex-matched healthy control subjects. We further performed haemostasis and platelet depletion tests in the R6/2 HD mouse model. RESULTS Our findings indicate that the platelets in HD are dysfunctional with respect to the release of angiogenic factors and functions including thrombosis, angiogenesis and vascular haemostasis. CONCLUSION Taken together, our results provide a better understanding for the impact of mHtt on platelet function.
Collapse
Affiliation(s)
- Hélèna L Denis
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Jérôme Lamontagne-Proulx
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Isabelle St-Amour
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Sarah L Mason
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Jesse W Rowley
- The Molecular Medicine Program and Department of Internal Medicine, University of Utah The George E. Wahlen VAMC GRECC, Salt Lake City, Utah, USA
| | - Nathalie Cloutier
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de microbiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de médecine moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Antony T Vincent
- Département de biochimie, de microbiologie et de bio-informatique, Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, QC, Canada
| | - Peter V Gould
- Département d'Anatomopathologie et de Cytologie, Centre Hospitalier Affilié Universitaire de Québec, Hôpital de l'Enfant-Jésus, Québec, QC, Canada
| | - Sylvain Chouinard
- Department of Movement Disorders, Centre Hospitalier Universitaire de Montréal-Hôtel Dieu, CHUM, Montréal, QC, Canada
| | - Andrew S Weyrich
- The Molecular Medicine Program and Department of Internal Medicine, University of Utah The George E. Wahlen VAMC GRECC, Salt Lake City, Utah, USA
| | - Matthew T Rondina
- The Molecular Medicine Program and Department of Internal Medicine, University of Utah The George E. Wahlen VAMC GRECC, Salt Lake City, Utah, USA
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Eric Boilard
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de microbiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| |
Collapse
|
39
|
Peteri UK, Niukkanen M, Castrén ML. Astrocytes in Neuropathologies Affecting the Frontal Cortex. Front Cell Neurosci 2019; 13:44. [PMID: 30809131 PMCID: PMC6379461 DOI: 10.3389/fncel.2019.00044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/28/2019] [Indexed: 01/15/2023] Open
Abstract
To an increasing extent, astrocytes are connected with various neuropathologies. Astrocytes comprise of a heterogeneous population of cells with region- and species-specific properties. The frontal cortex exhibits high levels of plasticity that is required for high cognitive functions and memory making this region especially susceptible to damage. Aberrations in the frontal cortex are involved with several cognitive disorders, including Alzheimer’s disease, Huntington’s disease and frontotemporal dementia. Human induced pluripotent stem cells (iPSCs) provide an alternative for disease modeling and offer possibilities for studies to investigate pathological mechanisms in a cell type-specific manner. Patient-specific iPSC-derived astrocytes have been shown to recapitulate several disease phenotypes. Addressing astrocyte heterogeneity may provide an improved understanding of the mechanisms underlying neurodegenerative diseases.
Collapse
Affiliation(s)
- Ulla-Kaisa Peteri
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Niukkanen
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maija L Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
40
|
Singh-Bains MK, Mehrabi NF, Sehji T, Austria MDR, Tan AYS, Tippett LJ, Dragunow M, Waldvogel HJ, Faull RLM. Cerebellar degeneration correlates with motor symptoms in Huntington disease. Ann Neurol 2019; 85:396-405. [PMID: 30635944 PMCID: PMC6590792 DOI: 10.1002/ana.25413] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Huntington disease (HD) is an autosomal dominant neurodegenerative disorder characterized by variable motor and behavioral symptoms attributed to major neuropathology of mainly the basal ganglia and cerebral cortex. The role of the cerebellum, a brain region involved in the coordination of movements, in HD neuropathology has been controversial. This study utilizes postmortem human brain tissue to investigate whether Purkinje cell degeneration in the neocerebellum is present in HD, and how this relates to disease symptom profiles. METHODS Unbiased stereological counting methods were used to quantify the total number of Purkinje cells in 15 HD cases and 8 neurologically normal control cases. Based on their predominant symptoms, the HD cases were categorized into 2 groups: "motor" or "mood." RESULTS The results demonstrated a significant 43% loss of Purkinje cells in HD cases with predominantly motor symptoms, and no cell loss in cases showing a major mood phenotype. There was no significant correlation between Purkinje cell loss and striatal neuropathological grade, postmortem delay, CAG repeat in the IT15 gene, or age at death. INTERPRETATION This study shows a compelling relationship between Purkinje cell loss in the HD neocerebellum and the HD motor symptom phenotype, which, together with our previous human brain studies on the same HD cases, provides novel perspectives interrelating and correlating the variable cerebellar, basal ganglia, and neocortical neuropathology with the variability of motor/mood symptom profiles in the human HD brain. ANN NEUROL 2019;85:396-405.
Collapse
Affiliation(s)
- Malvindar K Singh-Bains
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Nasim F Mehrabi
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Tvesa Sehji
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Micah D R Austria
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Adelie Y S Tan
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Lynette J Tippett
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Psychology, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| |
Collapse
|
41
|
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 2019; 99:21-78. [PMID: 30280653 PMCID: PMC6335099 DOI: 10.1152/physrev.00050.2017] [Citation(s) in RCA: 1301] [Impact Index Per Article: 216.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) prevents neurotoxic plasma components, blood cells, and pathogens from entering the brain. At the same time, the BBB regulates transport of molecules into and out of the central nervous system (CNS), which maintains tightly controlled chemical composition of the neuronal milieu that is required for proper neuronal functioning. In this review, we first examine molecular and cellular mechanisms underlying the establishment of the BBB. Then, we focus on BBB transport physiology, endothelial and pericyte transporters, and perivascular and paravascular transport. Next, we discuss rare human monogenic neurological disorders with the primary genetic defect in BBB-associated cells demonstrating the link between BBB breakdown and neurodegeneration. Then, we review the effects of genes underlying inheritance and/or increased susceptibility for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and amyotrophic lateral sclerosis (ALS) on BBB in relation to other pathologies and neurological deficits. We next examine how BBB dysfunction relates to neurological deficits and other pathologies in the majority of sporadic AD, PD, and ALS cases, multiple sclerosis, other neurodegenerative disorders, and acute CNS disorders such as stroke, traumatic brain injury, spinal cord injury, and epilepsy. Lastly, we discuss BBB-based therapeutic opportunities. We conclude with lessons learned and future directions, with emphasis on technological advances to investigate the BBB functions in the living human brain, and at the molecular and cellular level, and address key unanswered questions.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
42
|
Page S, Patel R, Raut S, Al-Ahmad A. Neurological diseases at the blood-brain barrier: Stemming new scientific paradigms using patient-derived induced pluripotent cells. Biochim Biophys Acta Mol Basis Dis 2018; 1866:165358. [PMID: 30593893 DOI: 10.1016/j.bbadis.2018.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 02/08/2023]
Abstract
The blood-brain barrier (BBB) is a component of the neurovascular unit formed by specialized brain microvascular endothelial cells (BMECs) surrounded by a specific basement membrane interacting with astrocytes, neurons, and pericytes. The BBB plays an essential function in the maintenance of brain homeostasis, by providing a physical and chemical barrier against pathogens and xenobiotics. Although the disruption of the BBB occurs with several neurological disorders, the scarcity of patient material source and lack of reliability of current in vitro models hindered our ability to model the BBB during such neurological conditions. The development of novel in vitro models based on patient-derived stem cells opened new venues in modeling the human BBB in vitro, by being more accurate than existing in vitro models, but also bringing such models closer to the in vivo setting. In addition, patient-derived models of the BBB opens the avenue to address the contribution of genetic factors commonly associated with certain neurological diseases on the BBB pathophysiology. This review provides a comprehensive understanding of the BBB, the current development of stem cell-based models in the field, the current challenges and limitations of such models.
Collapse
Affiliation(s)
- Shyanne Page
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Ronak Patel
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Snehal Raut
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX, United States of America.
| |
Collapse
|
43
|
Bordoni M, Rey F, Fantini V, Pansarasa O, Di Giulio AM, Carelli S, Cereda C. From Neuronal Differentiation of iPSCs to 3D Neuro-Organoids: Modelling and Therapy of Neurodegenerative Diseases. Int J Mol Sci 2018; 19:E3972. [PMID: 30544711 PMCID: PMC6321164 DOI: 10.3390/ijms19123972] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/27/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the advances made into the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) led to great improvements towards their use as models of diseases. In particular, in the field of neurodegenerative diseases, iPSCs technology allowed to culture in vitro all types of patient-specific neural cells, facilitating not only the investigation of diseases' etiopathology, but also the testing of new drugs and cell therapies, leading to the innovative concept of personalized medicine. Moreover, iPSCs can be differentiated and organized into 3D organoids, providing a tool which mimics the complexity of the brain's architecture. Furthermore, recent developments in 3D bioprinting allowed the study of physiological cell-to-cell interactions, given by a combination of several biomaterials, scaffolds, and cells. This technology combines bio-plotter and biomaterials in which several types of cells, such as iPSCs or differentiated neurons, can be encapsulated in order to develop an innovative cellular model. IPSCs and 3D cell cultures technologies represent the first step towards the obtainment of a more reliable model, such as organoids, to facilitate neurodegenerative diseases' investigation. The combination of iPSCs, 3D organoids and bioprinting will also allow the development of new therapeutic approaches. Indeed, on the one hand they will lead to the development of safer and patient-specific drugs testing but, also, they could be developed as cell-therapy for curing neurodegenerative diseases with a regenerative medicine approach.
Collapse
Affiliation(s)
- Matteo Bordoni
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Federica Rey
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142 Milan, Italy.
| | - Valentina Fantini
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy.
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy.
| | - Orietta Pansarasa
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Anna Maria Di Giulio
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142 Milan, Italy.
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of MilanVia Giovanni Battista Grassi, 74, 20157 Milan, Italy.
| | - Stephana Carelli
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, via A. di Rudinì 8, 20142 Milan, Italy.
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of MilanVia Giovanni Battista Grassi, 74, 20157 Milan, Italy.
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy.
| |
Collapse
|
44
|
Chang WT, Puspitasari F, Garcia-Miralles M, Yeow LY, Tay HC, Koh KB, Tan LJ, Pouladi MA, Chuang KH. Connectomic imaging reveals Huntington-related pathological and pharmaceutical effects in a mouse model. NMR IN BIOMEDICINE 2018; 31:e4007. [PMID: 30260561 DOI: 10.1002/nbm.4007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/05/2018] [Accepted: 07/31/2018] [Indexed: 06/08/2023]
Abstract
Recent studies suggest that neurodegenerative diseases could affect brain structure and function in disease-specific network patterns; however, how spontaneous activity affects structural covariance network (SC) is not clear. We hypothesized that hyper-excitability in Huntington disease (HD) disrupts the coordinated structural and functional connectivity, and treatment with memantine helps to reduce excitotoxicity and normalize the connectivity. MRI was conducted to measure somatosensory activation, resting-state functional-connectivity (rsFC), SC, amplitude of low frequency fluctuation (ALFF) and ALFF covariance (ALFFC) in the YAC128 mouse model of HD. We found somatosensory activation was unchanged but the subcortical ALFF was increased in HD mice, indicating subcortical but not cortical hyperactivity. The reduced sensorimotor rsFC but spared hippocampal and default mode networks in the HD mice was consistent with the more pronounced impairment in motor function compared with cognitive performance. The disease suppressed SC globally and reduced ALFFC in the basal ganglia network as well as its anti-correlation with the default mode network. By comparing these connectivity measures, we found that the originally coupled rsFC-SC relationship was impaired whereas SC-ALFFC correlation was increased by HD, suggesting disease facilitated covariation of brain volume and activity amplitude but not neural synchrony. The comparison with mono-synaptic axonal projection supports the hypothesis that rsFC, but not SC or ALFFC, is highly dependent on structural connectivity under healthy conditions. Treatment with memantine had a strong effect on normalizing the SC and reducing ALFF while slightly increasing other connectivity measures and restoring the rsFC-SC coupling, which is consistent with its effect on alleviating hyper-excitability and improving the coordinated neural growth. These results indicate that HD affects the cerebral structure-function relationship which could be partially reverted by NMDA antagonism. These connectivity measures provide unique insights into pathological and pharmaceutical effects in brain circuitry, and could be translatable biomarkers for evaluating drug effect and refining its efficacy.
Collapse
Affiliation(s)
- Wei-Tang Chang
- Singapore BioImaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
| | - Fiftarina Puspitasari
- Singapore BioImaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
| | - Marta Garcia-Miralles
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ling Yun Yeow
- Singapore BioImaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hui-Chien Tay
- Singapore BioImaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
| | - Katrianne Bethia Koh
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore, Singapore
| | - Liang Juin Tan
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore, Singapore
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai-Hsiang Chuang
- Singapore BioImaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| |
Collapse
|
45
|
Trias E, Barbeito L, Yamanaka K. Phenotypic heterogeneity of astrocytes in motor neuron disease. ACTA ACUST UNITED AC 2018; 9:225-234. [PMID: 30555538 PMCID: PMC6282976 DOI: 10.1111/cen3.12476] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/22/2018] [Indexed: 12/20/2022]
Abstract
Accumulating evidence has shown that astrocytes do not just support the function of neurons, but play key roles in maintaining the brain environment in health and disease. Contrary to the traditional understanding of astrocytes as static cells, reactive astrocytes possess more diverse functions and phenotypes than previously predicted. In the present focused review, we summarize the evidence showing that astrocytes are playing profound roles in the disease process of amyotrophic lateral sclerosis. Aberrantly activated astrocytes in amyotrophic lateral sclerosis rodents express microglial molecular markers and provoke toxicities to accelerate disease progression. In addition, TIR domain–containing adapter protein–inducing interferon‐β‐dependent innate immune pathway in astrocytes also has a novel function in terminating glial activation and neuroinflammation. Furthermore, heterogeneity in phenotypes and functions of astrocytes are also observed in various disease conditions, such as other neurodegenerative diseases, ischemia, aging and acute lesions in the central nervous system. Through accumulating knowledge of the phenotypic and functional diversity of astrocytes, these cells will become more attractive therapeutic targets for neurological diseases.
Collapse
Affiliation(s)
| | | | - Koji Yamanaka
- Department of Neuroscience and Pathobiology Research Institute of Environmental Medicine Nagoya University Nagoya Japan.,Department of Neuroscience and Pathobiology Nagoya University Graduate School of Medicine Nagoya Japan
| |
Collapse
|
46
|
Cheng J, Korte N, Nortley R, Sethi H, Tang Y, Attwell D. Targeting pericytes for therapeutic approaches to neurological disorders. Acta Neuropathol 2018; 136:507-523. [PMID: 30097696 PMCID: PMC6132947 DOI: 10.1007/s00401-018-1893-0] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022]
Abstract
Many central nervous system diseases currently lack effective treatment and are often associated with defects in microvascular function, including a failure to match the energy supplied by the blood to the energy used on neuronal computation, or a breakdown of the blood–brain barrier. Pericytes, an under-studied cell type located on capillaries, are of crucial importance in regulating diverse microvascular functions, such as angiogenesis, the blood–brain barrier, capillary blood flow and the movement of immune cells into the brain. They also form part of the “glial” scar isolating damaged parts of the CNS, and may have stem cell-like properties. Recent studies have suggested that pericytes play a crucial role in neurological diseases, and are thus a therapeutic target in disorders as diverse as stroke, traumatic brain injury, migraine, epilepsy, spinal cord injury, diabetes, Huntington’s disease, Alzheimer’s disease, diabetes, multiple sclerosis, glioma, radiation necrosis and amyotrophic lateral sclerosis. Here we report recent advances in our understanding of pericyte biology and discuss how pericytes could be targeted to develop novel therapeutic approaches to neurological disorders, by increasing blood flow, preserving blood–brain barrier function, regulating immune cell entry to the CNS, and modulating formation of blood vessels in, and the glial scar around, damaged regions.
Collapse
Affiliation(s)
- Jinping Cheng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Rd, Guangzhou, 510120, People's Republic of China
| | - Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ross Nortley
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Huma Sethi
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Rd, Guangzhou, 510120, People's Republic of China.
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
47
|
Joshi AU, Mochly-Rosen D. Mortal engines: Mitochondrial bioenergetics and dysfunction in neurodegenerative diseases. Pharmacol Res 2018; 138:2-15. [PMID: 30144530 DOI: 10.1016/j.phrs.2018.08.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022]
Abstract
Mitochondria are best known for their role in ATP generation. However, studies over the past two decades have shown that mitochondria do much more than that. Mitochondria regulate both necrotic and apoptotic cell death pathways, they store and therefore coordinate cellular Ca2+ signaling, they generate and metabolize important building blocks, by-products and signaling molecules, and they also generate and are targets of free radical species that modulate many aspects of cell physiology and pathology. Most estimates suggest that although the brain makes up only 2 percent of body weight, utilizes about 20 percent of the body's total ATP. Thus, mitochondrial dysfunction greatly impacts brain functions and is indeed associated with numerous neurodegenerative diseases. Furthermore, a number of abnormal disease-associated proteins have been shown to interact directly with mitochondria, leading to mitochondrial dysfunction and subsequent neuronal cell death. Here, we discuss the role of mitochondrial dynamics impairment in the pathological processes associated with neurodegeneration and suggest that a therapy targeting mitochondrialdysfunction holds a great promise.
Collapse
Affiliation(s)
- Amit U Joshi
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, CA, 94305-5174, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, CA, 94305-5174, USA.
| |
Collapse
|
48
|
Watts ME, Pocock R, Claudianos C. Brain Energy and Oxygen Metabolism: Emerging Role in Normal Function and Disease. Front Mol Neurosci 2018; 11:216. [PMID: 29988368 PMCID: PMC6023993 DOI: 10.3389/fnmol.2018.00216] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/01/2018] [Indexed: 01/09/2023] Open
Abstract
Dynamic metabolic changes occurring in neurons are critically important in directing brain plasticity and cognitive function. In other tissue types, disruptions to metabolism and the resultant changes in cellular oxidative state, such as increased reactive oxygen species (ROS) or induction of hypoxia, are associated with cellular stress. In the brain however, where drastic metabolic shifts occur to support physiological processes, subsequent changes to cellular oxidative state and induction of transcriptional sensors of oxidative stress likely play a significant role in regulating physiological neuronal function. Understanding the role of metabolism and metabolically-regulated genes in neuronal function will be critical in elucidating how cognitive functions are disrupted in pathological conditions where neuronal metabolism is affected. Here, we discuss known mechanisms regulating neuronal metabolism as well as the role of hypoxia and oxidative stress during normal and disrupted neuronal function. We also summarize recent studies implicating a role for metabolism in regulating neuronal plasticity as an emerging neuroscience paradigm.
Collapse
Affiliation(s)
- Michelle E Watts
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Charles Claudianos
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia.,Centre for Mental Health Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
49
|
Padel T, Roth M, Gaceb A, Li JY, Björkqvist M, Paul G. Brain pericyte activation occurs early in Huntington's disease. Exp Neurol 2018; 305:139-150. [PMID: 29630897 DOI: 10.1016/j.expneurol.2018.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/03/2018] [Accepted: 03/29/2018] [Indexed: 02/03/2023]
Abstract
Microvascular changes have recently been described for several neurodegenerative disorders, including Huntington's disease (HD). HD is characterized by a progressive neuronal cell loss due to a mutation in the Huntingtin gene. However, the temporal and spatial microvascular alterations in HD remain unclear. Also, knowledge on the implication of pericytes in HD pathology is still sparse and existing findings are contradictory. Here we examine alterations in brain pericytes in the R6/2 mouse model of HD and in human post mortem HD brain sections. To specifically track activated pericytes, we crossbred R6/2 mice with transgenic mice expressing the Green fluorescent protein gene under the Regulator of G-protein signaling 5 (Rgs5) promoter. We demonstrate an increase in activated pericytes in the R6/2 brain and in post mortem HD brain tissue. Importantly, pericyte changes are already detected before striatal neuronal cell loss, weight loss or behavioural deficits occur in R6/2 mice. This is associated with vascular alterations, whereby striatal changes precede cortical changes. Our findings suggest that pericyte activation may be one of the initial steps contributing to the observed vascular modifications in HD. Thus, pericytes may constitute an important target to address early microvascular changes contributing to disease progression in HD.
Collapse
Affiliation(s)
- Thomas Padel
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Michaela Roth
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden; Department of Neurology, Scania University Hospital, 22185 Lund, Sweden.
| |
Collapse
|
50
|
Lim RG, Quan C, Reyes-Ortiz AM, Lutz SE, Kedaigle AJ, Gipson TA, Wu J, Vatine GD, Stocksdale J, Casale MS, Svendsen CN, Fraenkel E, Housman DE, Agalliu D, Thompson LM. Huntington's Disease iPSC-Derived Brain Microvascular Endothelial Cells Reveal WNT-Mediated Angiogenic and Blood-Brain Barrier Deficits. Cell Rep 2018; 19:1365-1377. [PMID: 28514657 PMCID: PMC5646270 DOI: 10.1016/j.celrep.2017.04.021] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 03/08/2017] [Accepted: 04/06/2017] [Indexed: 12/16/2022] Open
Abstract
Brain microvascular endothelial cells (BMECs) are an essential component of the blood-brain barrier (BBB) that shields the brain against toxins and immune cells. While BBB dysfunction exists in neurological disorders, including Huntington’s disease (HD), it is not known if BMECs themselves are functionally compromised to promote BBB dysfunction. Further, the underlying mechanisms of BBB dysfunction remain elusive given limitations with mouse models and post-mortem tissue to identify primary deficits. We undertook a transcriptome and functional analysis of human induced pluripotent stem cell (iPSC)-derived BMECs (iBMEC) from HD patients or unaffected controls. We demonstrate that HD iBMECs have intrinsic abnormalities in angiogenesis and barrier properties, as well as in signaling pathways governing these processes. Thus, our findings provide an iPSC-derived BBB model for a neurodegenerative disease and demonstrate autonomous neurovascular deficits that may underlie HD pathology with implications for therapeutics and drug delivery.
Collapse
Affiliation(s)
- Ryan G Lim
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; UCI MIND, University of California, Irvine, Irvine, CA 92697, USA
| | - Chris Quan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Boulevard, Long Beach, CA 90840, USA
| | - Andrea M Reyes-Ortiz
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Sarah E Lutz
- Departments of Neurology, Pathology, and Cell Biology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Amanda J Kedaigle
- Computational and Systems Biology Graduate Program, MIT, Cambridge, MA 02139, USA
| | - Theresa A Gipson
- Center for Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Gad D Vatine
- Department of Biomedical Sciences, The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Malcolm S Casale
- Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Clive N Svendsen
- Department of Biomedical Sciences, The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - David E Housman
- Center for Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Dritan Agalliu
- Departments of Neurology, Pathology, and Cell Biology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA; Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY 10032, USA.
| | - Leslie M Thompson
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; UCI MIND, University of California, Irvine, Irvine, CA 92697, USA; Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|