1
|
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
|
2
|
Tzoneva R. Special Issue "Role of Apoptosis and Cellular Senescence in Cancer and Aging". Int J Mol Sci 2024; 25:2103. [PMID: 38396780 PMCID: PMC10889768 DOI: 10.3390/ijms25042103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The intention of this Special Issue is to elucidate the role of apoptosis and cellular senescence in different pathological processes, such as cancer and aging [...].
Collapse
Affiliation(s)
- Rumiana Tzoneva
- Laboratory of Transmembrane Signaling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Akad. G. Bonchev Str., bl. 21, 1113 Sofia, Bulgaria
| |
Collapse
|
3
|
Georgieva I, Tchekalarova J, Iliev D, Tzoneva R. Endothelial Senescence and Its Impact on Angiogenesis in Alzheimer's Disease. Int J Mol Sci 2023; 24:11344. [PMID: 37511104 PMCID: PMC10379128 DOI: 10.3390/ijms241411344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Endothelial cells are constantly exposed to environmental stress factors that, above a certain threshold, trigger cellular senescence and apoptosis. The altered vascular function affects new vessel formation and endothelial fitness, contributing to the progression of age-related diseases. This narrative review highlights the complex interplay between senescence, oxidative stress, extracellular vesicles, and the extracellular matrix and emphasizes the crucial role of angiogenesis in aging and Alzheimer's disease. The interaction between the vascular and nervous systems is essential for the development of a healthy brain, especially since neurons are exceptionally dependent on nutrients carried by the blood. Therefore, anomalies in the delicate balance between pro- and antiangiogenic factors and the consequences of disrupted angiogenesis, such as misalignment, vascular leakage and disturbed blood flow, are responsible for neurodegeneration. The implications of altered non-productive angiogenesis in Alzheimer's disease due to dysregulated Delta-Notch and VEGF signaling are further explored. Additionally, potential therapeutic strategies such as exercise and caloric restriction to modulate angiogenesis and vascular aging and to mitigate the associated debilitating symptoms are discussed. Moreover, both the roles of extracellular vesicles in stress-induced senescence and as an early detection marker for Alzheimer's disease are considered. The intricate relationship between endothelial senescence and angiogenesis provides valuable insights into the mechanisms underlying angiogenesis-related disorders and opens avenues for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Irina Georgieva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. George Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria
| | - Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. George Bonchev, Str. Bl. 23, 1113 Sofia, Bulgaria
| | - Dimitar Iliev
- Institute of Molecular Biology, Bulgarian Academy of Sciences, Acad. George Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria
| | - Rumiana Tzoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. George Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria
| |
Collapse
|
4
|
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
|
5
|
Gupta R, Ambasta RK, Pravir Kumar. Autophagy and apoptosis cascade: which is more prominent in neuronal death? Cell Mol Life Sci 2021; 78:8001-8047. [PMID: 34741624 PMCID: PMC11072037 DOI: 10.1007/s00018-021-04004-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
Autophagy and apoptosis are two crucial self-destructive processes that maintain cellular homeostasis, which are characterized by their morphology and regulated through signal transduction mechanisms. These pathways determine the fate of cellular organelle and protein involved in human health and disease such as neurodegeneration, cancer, and cardiovascular disease. Cell death pathways share common molecular mechanisms, such as mitochondrial dysfunction, oxidative stress, calcium ion concentration, reactive oxygen species, and endoplasmic reticulum stress. Some key signaling molecules such as p53 and VEGF mediated angiogenic pathway exhibit cellular and molecular responses resulting in the triggering of apoptotic and autophagic pathways. Herein, based on previous studies, we describe the intricate relation between cell death pathways through their common genes and the role of various stress-causing agents. Further, extensive research on autophagy and apoptotic machinery excavates the implementation of selective biomarkers, for instance, mTOR, Bcl-2, BH3 family members, caspases, AMPK, PI3K/Akt/GSK3β, and p38/JNK/MAPK, in the pathogenesis and progression of neurodegenerative diseases. This molecular phenomenon will lead to the discovery of possible therapeutic biomolecules as a pharmacological intervention that are involved in the modulation of apoptosis and autophagy pathways. Moreover, we describe the potential role of micro-RNAs, long non-coding RNAs, and biomolecules as therapeutic agents that regulate cell death machinery to treat neurodegenerative diseases. Mounting evidence demonstrated that under stress conditions, such as calcium efflux, endoplasmic reticulum stress, the ubiquitin-proteasome system, and oxidative stress intermediate molecules, namely p53 and VEGF, activate and cause cell death. Further, activation of p53 and VEGF cause alteration in gene expression and dysregulated signaling pathways through the involvement of signaling molecules, namely mTOR, Bcl-2, BH3, AMPK, MAPK, JNK, and PI3K/Akt, and caspases. Alteration in gene expression and signaling cascades cause neurotoxicity and misfolded protein aggregates, which are characteristics features of neurodegenerative diseases. Excessive neurotoxicity and misfolded protein aggregates lead to neuronal cell death by activating death pathways like autophagy and apoptosis. However, autophagy has a dual role in the apoptosis pathways, i.e., activation and inhibition of the apoptosis signaling. Further, micro-RNAs and LncRNAs act as pharmacological regulators of autophagy and apoptosis cascade, whereas, natural compounds and chemical compounds act as pharmacological inhibitors that rescue neuronal cell death through inhibition of apoptosis and autophagic cell death.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
- , Delhi, India.
| |
Collapse
|
6
|
Wang X, Ma C, Rodríguez Labrada R, Qin Z, Xu T, He Z, Wei Y. Recent advances in lentiviral vectors for gene therapy. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1842-1857. [PMID: 34708326 DOI: 10.1007/s11427-021-1952-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023]
Abstract
Lentiviral vectors (LVs), derived from human immunodeficiency virus, are powerful tools for modifying the genes of eukaryotic cells such as hematopoietic stem cells and neural cells. With the extensive and in-depth studies on this gene therapy vehicle over the past two decades, LVs have been widely used in both research and clinical trials. For instance, third-generation and self-inactive LVs have been used to introduce a gene with therapeutic potential into the host genome and achieve targeted delivery into specific tissue. When LVs are employed in leukemia, the transduced T cells recognize and kill the tumor B cells; in β-thalassemia, the transduced CD34+ cells express normal β-globin; in adenosine deaminase-deficient severe combined immunodeficiency, the autologous CD34+ cells express adenosine deaminase and realize immune reconstitution. Overall, LVs can perform significant roles in the treatment of primary immunodeficiency diseases, hemoglobinopathies, B cell leukemia, and neurodegenerative diseases. In this review, we discuss the recent developments and therapeutic applications of LVs. The safe and efficient LVs show great promise as a tool for human gene therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cuicui Ma
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Roberto Rodríguez Labrada
- Department Clinical Neurophysiology, Centre for the Research and Rehabilitation of Hereditary Ataxias, Holguín, 80100, Cuba
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Yuquan Wei
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Kim A, Lalonde K, Truesdell A, Gomes Welter P, Brocardo PS, Rosenstock TR, Gil-Mohapel J. New Avenues for the Treatment of Huntington's Disease. Int J Mol Sci 2021; 22:ijms22168363. [PMID: 34445070 PMCID: PMC8394361 DOI: 10.3390/ijms22168363] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HD gene. The disease is characterized by neurodegeneration, particularly in the striatum and cortex. The first symptoms usually appear in mid-life and include cognitive deficits and motor disturbances that progress over time. Despite being a genetic disorder with a known cause, several mechanisms are thought to contribute to neurodegeneration in HD, and numerous pre-clinical and clinical studies have been conducted and are currently underway to test the efficacy of therapeutic approaches targeting some of these mechanisms with varying degrees of success. Although current clinical trials may lead to the identification or refinement of treatments that are likely to improve the quality of life of those living with HD, major efforts continue to be invested at the pre-clinical level, with numerous studies testing novel approaches that show promise as disease-modifying strategies. This review offers a detailed overview of the currently approved treatment options for HD and the clinical trials for this neurodegenerative disorder that are underway and concludes by discussing potential disease-modifying treatments that have shown promise in pre-clinical studies, including increasing neurotropic support, modulating autophagy, epigenetic and genetic manipulations, and the use of nanocarriers and stem cells.
Collapse
Affiliation(s)
- Amy Kim
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Kathryn Lalonde
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Aaron Truesdell
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Priscilla Gomes Welter
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Patricia S. Brocardo
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Tatiana R. Rosenstock
- Institute of Cancer and Genomic Science, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Department of Pharmacology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Joana Gil-Mohapel
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Correspondence: ; Tel.: +1-250-472-4597; Fax: +1-250-472-5505
| |
Collapse
|
8
|
Prados ME, Correa-Sáez A, Unciti-Broceta JD, Garrido-Rodríguez M, Jimenez-Jimenez C, Mazzone M, Minassi A, Appendino G, Calzado MA, Muñoz E. Betulinic Acid Hydroxamate is Neuroprotective and Induces Protein Phosphatase 2A-Dependent HIF-1α Stabilization and Post-transcriptional Dephosphorylation of Prolyl Hydrolase 2. Neurotherapeutics 2021; 18:1849-1861. [PMID: 34339019 PMCID: PMC8608974 DOI: 10.1007/s13311-021-01089-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 02/04/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by unwanted choreatic movements, behavioral and psychiatric disturbances, and dementia. The activation of the hypoxic response pathway through the pharmacological inhibition of hypoxia-inducing factor (HIF) prolyl-hydroxylases (PHDs) is a promising approach for neurodegenerative diseases, including HD. Herein, we have studied the mechanism of action of the compound Betulinic acid hydroxamate (BAH), a hypoximimetic derivative of betulinic acid, and its efficacy against striatal neurodegeneration using complementary approaches. Firstly, we showed the molecular mechanisms through which BAH modifies the activity of the PHD2 prolyl hydroxylase, thus directly affecting HIF-1α stability. BAH treatment reduces PHD2 phosphorylation on Ser-125 residue, responsible for the control of its hydrolase activity. HIF activation by BAH is inhibited by okadaic acid and LB-100 indicating that a protein phosphatase 2A (PP2A) is implicated in the mechanism of action of BAH. Furthermore, in striatal cells bearing a mutated form of the huntingtin protein, BAH stabilized HIF-1α protein, induced Vegf and Bnip3 gene expression and protected against mitochondrial toxin-induced cytotoxicity. Pharmacokinetic analyses showed that BAH has a good brain penetrability and experiments performed in a mouse model of striatal neurodegeneration induced by 3-nitropropionic acid showed that BAH improved the clinical symptoms. In addition, BAH also prevented neuronal loss, decreased reactive astrogliosis and microglial activation, inhibited the upregulation of proinflammatory markers, and improved antioxidant defenses in the brain. Taken together, our results show BAH's ability to activate the PP2A/PHD2/HIF pathway, which may have important implications in the treatment of HD and perhaps other neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Alejandro Correa-Sáez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
| | | | - Martín Garrido-Rodríguez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
| | - Carla Jimenez-Jimenez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB-KULeuven, 3000, Leuven, Belgium
| | - Alberto Minassi
- Department of Drug Science, University of Piemonte Orientale, Novara, Italy
| | - Giovanni Appendino
- Department of Drug Science, University of Piemonte Orientale, Novara, Italy
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain.
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain.
| |
Collapse
|
9
|
Červenka J, Tylečková J, Kupcová Skalníková H, Vodičková Kepková K, Poliakh I, Valeková I, Pfeiferová L, Kolář M, Vaškovičová M, Pánková T, Vodička P. Proteomic Characterization of Human Neural Stem Cells and Their Secretome During in vitro Differentiation. Front Cell Neurosci 2021; 14:612560. [PMID: 33584205 PMCID: PMC7876319 DOI: 10.3389/fncel.2020.612560] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
Cell therapies represent a promising approach to slow down the progression of currently untreatable neurodegenerative diseases (e.g., Alzheimer's and Parkinson's disease or amyotrophic lateral sclerosis), as well as to support the reconstruction of functional neural circuits after spinal cord injuries. In such therapies, the grafted cells could either functionally integrate into the damaged tissue, partially replacing dead or damaged cells, modulate inflammatory reaction, reduce tissue damage, or support neuronal survival by secretion of cytokines, growth, and trophic factors. Comprehensive characterization of cells and their proliferative potential, differentiation status, and population purity before transplantation is crucial to preventing safety risks, e.g., a tumorous growth due to the proliferation of undifferentiated stem cells. We characterized changes in the proteome and secretome of human neural stem cells (NSCs) during their spontaneous (EGF/FGF2 withdrawal) differentiation and differentiation with trophic support by BDNF/GDNF supplementation. We used LC-MS/MS in SWATH-MS mode for global cellular proteome profiling and quantified almost three thousand cellular proteins. Our analysis identified substantial protein differences in the early stages of NSC differentiation with more than a third of all the proteins regulated (including known neuronal and NSC multipotency markers) and revealed that the BDNF/GDNF support affected more the later stages of the NSC differentiation. Among the pathways identified as activated during both spontaneous and BDNF/GDNF differentiation were the HIF-1 signaling pathway, Wnt signaling pathway, and VEGF signaling pathway. Our follow-up secretome analysis using Luminex multiplex immunoassay revealed significant changes in the secretion of VEGF and IL-6 during NSC differentiation. Our results further demonstrated an increased expression of neuropilin-1 as well as catenin β-1, both known to participate in the regulation of VEGF signaling, and showed that VEGF-A isoform 121 (VEGF121), in particular, induces proliferation and supports survival of differentiating cells.
Collapse
Affiliation(s)
- Jakub Červenka
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia.,Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
| | - Jiřina Tylečková
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| | - Helena Kupcová Skalníková
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| | - Kateřina Vodičková Kepková
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| | - Ievgeniia Poliakh
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia.,Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
| | - Ivona Valeková
- Laboratory of Cell Regeneration and Plasticity, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| | - Lucie Pfeiferová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia.,Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague, Czechia
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Michaela Vaškovičová
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia.,Laboratory of DNA Integrity, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| | - Tereza Pánková
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia.,Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
| | - Petr Vodička
- Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czechia
| |
Collapse
|
10
|
Eskandari N, Boroujeni ME, Abdollahifar MA, Piryaei A, Khodagholi F, Mirbehbahani SH, Siroosi S, Moghaddam MH, Aliaghaei A, Sadeghi Y. Transplantation of human dental pulp stem cells compensates for striatal atrophy and modulates neuro-inflammation in 3-nitropropionic acid rat model of Huntington's disease. Neurosci Res 2020; 170:133-144. [PMID: 33359180 DOI: 10.1016/j.neures.2020.12.002] [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: 09/03/2020] [Revised: 11/09/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023]
Abstract
Stem cell-based therapy has recently offered a promising alternative for the remedy of neurodegenerative disorders like Huntington's disease (HD). Herein, we investigated the potential ameliorative effects of implantation of dental pulp stem cells (DPSCs) in 3-nitropropionic acid (3-NP) rat models of HD. In this regard, human DPSCs were isolated, culture-expanded and implanted in rats lesioned with 3-NP. Post-transplantation examinations revealed that DPSCs were able to survive and augment motor skills and muscle activity. Histological analysis showed DPSCs treatment hampered the shrinkage of the striatum along with the inhibition of gliosis and microgliosis in the striatum of 3-NP rat models. We also detected the downregulation of Caspase-3 and pro-inflammatory cytokines such as TNF and IL-1β upon DPSCs grafting. Overall, these findings imply that the grafting of DPSCs could repair motor-skill impairment and induce neurogenesis, probably through the secretion of neurotrophic factors and the modulation of neuroinflammatory response in HD animal models.
Collapse
Affiliation(s)
- Neda Eskandari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Mohammad Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shokoofeh Siroosi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Yousef Sadeghi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Anatomy & Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Denis HL, Lauruol F, Cicchetti F. Are immunotherapies for Huntington's disease a realistic option? Mol Psychiatry 2019; 24:364-377. [PMID: 29487401 DOI: 10.1038/s41380-018-0021-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/22/2017] [Accepted: 01/15/2018] [Indexed: 01/28/2023]
Abstract
There is compelling evidence that the pathophysiology of many neurodegenerative diseases includes dysregulation of the immune system, with some elements that precede disease onset. However, if these alterations are prominent, why have clinical trials targeting this system failed to translate into long-lasting meaningful benefits for patients? This review focuses on Huntington's disease, a genetic disorder marked by notable cerebral and peripheral inflammation. We summarize ongoing and completed clinical trials that have involved pharmacological approaches to inhibit various components of the immune system and their pre-clinical correlates. We then discuss new putative treatment strategies using more targeted immunotherapies such as vaccination and intrabodies and how these may offer new hope in the treatment of Huntington's disease as well as other neurodegenerative diseases.
Collapse
Affiliation(s)
- Hélèna L Denis
- Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, G1V 4G2, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Florian Lauruol
- Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, G1V 4G2, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, G1V 4G2, Canada. .,Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
12
|
Human Umbilical Cord Matrix Stem Cells Reverse Oxidative Stress-Induced Cell Death and Ameliorate Motor Function and Striatal Atrophy in Rat Model of Huntington Disease. Neurotox Res 2018. [PMID: 29520722 DOI: 10.1007/s12640-018-9884-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Eleftheriadou I, Dieringer M, Poh XY, Sanchez-Garrido J, Gao Y, Sgourou A, Simmons LE, Mazarakis ND. Selective transduction of astrocytic and neuronal CNS subpopulations by lentiviral vectors pseudotyped with Chikungunya virus envelope. Biomaterials 2017; 123:1-14. [PMID: 28152379 DOI: 10.1016/j.biomaterials.2017.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 12/15/2022]
Abstract
Lentiviral vectors are gene delivery vehicles that integrate into the host genome of dividing and non-dividing mammalian cells facilitating long-term transgene expression. Lentiviral vector versatility is greatly increased by incorporating heterologous viral envelope proteins onto the vector particles instead of the native envelope, conferring on these pseudotyped vectors a modified tropism and host range specificity. We investigated the pseudotyping efficiency of HIV-1 based lentiviral vectors with alphaviral envelope proteins from the Chikungunya Virus (CHIKV-G) and Sindbis Virus (SINV-G). Following vector production optimisation, titres for the CHIKV-G pseudotype were comparable to the VSV-G pseudotype but those for the SINV-G pseudotype were significantly lower. High titre CHIKV-G pseudotyped vector efficiently transduced various human and mouse neural cell lines and normal human astrocytes (NHA) in vitro. Although transduction was broad, tropism for NHAs was observed. In vivo stereotaxic delivery in striatum, thalamus and hippocampus respectively in the adult rat brain revealed localised transduction restricted to striatal astrocytes and hippocampal dentate granule neurons. Transduction of different subtypes of granule neurons from precursor to post-mitotic stages of differentiation was evident in the sub-granular zone and dentate granule cell layer. No significant inflammatory response was observed, but comparable to that of VSV-G pseudotyped lentiviral vectors. Robust long-term expression followed for three months post-transduction along with absence of neuroinflammation, coupled to the selective and unique neuron/glial tropism indicates that these vectors could be useful for modelling and gene therapy studies in the CNS.
Collapse
Affiliation(s)
- Ioanna Eleftheriadou
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom
| | - Michael Dieringer
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom
| | - Xuan Ying Poh
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom
| | - Julia Sanchez-Garrido
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom
| | - Yunan Gao
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom
| | - Argyro Sgourou
- Laboratory of Biology, Hellenic Open University, Tsamadou 13-15, 26222 Patra, Greece
| | - Laura E Simmons
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom
| | - Nicholas D Mazarakis
- Gene Therapy, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, United Kingdom.
| |
Collapse
|
14
|
Lange C, Storkebaum E, de Almodóvar CR, Dewerchin M, Carmeliet P. Vascular endothelial growth factor: a neurovascular target in neurological diseases. Nat Rev Neurol 2016; 12:439-54. [PMID: 27364743 DOI: 10.1038/nrneurol.2016.88] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain function critically relies on blood vessels to supply oxygen and nutrients, to establish a barrier for neurotoxic substances, and to clear waste products. The archetypal vascular endothelial growth factor, VEGF, arose in evolution as a signal affecting neural cells, but was later co-opted by blood vessels to regulate vascular function. Consequently, VEGF represents an attractive target to modulate brain function at the neurovascular interface. On the one hand, VEGF is neuroprotective, through direct effects on neural cells and their progenitors and indirect effects on brain perfusion. In accordance, preclinical studies show beneficial effects of VEGF administration in neurodegenerative diseases, peripheral neuropathies and epilepsy. On the other hand, pathologically elevated VEGF levels enhance vessel permeability and leakage, and disrupt blood-brain barrier integrity, as in demyelinating diseases, for which blockade of VEGF may be beneficial. Here, we summarize current knowledge on the role and therapeutic potential of VEGF in neurological diseases.
Collapse
Affiliation(s)
- Christian Lange
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| | - Erik Storkebaum
- Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149 Muenster, Germany.,Faculty of Medicine, University of Muenster, Roentgenstrasse 20, D-48149 Muenster, Germany
| | | | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| |
Collapse
|
15
|
Tovar-y-Romo LB, Penagos-Puig A, Ramírez-Jarquín JO. Endogenous recovery after brain damage: molecular mechanisms that balance neuronal life/death fate. J Neurochem 2015; 136:13-27. [PMID: 26376102 DOI: 10.1111/jnc.13362] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 07/27/2015] [Accepted: 08/25/2015] [Indexed: 01/08/2023]
Abstract
Neuronal survival depends on multiple factors that comprise a well-fueled energy metabolism, trophic input, clearance of toxic substances, appropriate redox environment, integrity of blood-brain barrier, suppression of programmed cell death pathways and cell cycle arrest. Disturbances of brain homeostasis lead to acute or chronic alterations that might ultimately cause neuronal death with consequent impairment of neurological function. Although we understand most of these processes well when they occur independently from one another, we still lack a clear grasp of the concerted cellular and molecular mechanisms activated upon neuronal damage that intervene in protecting damaged neurons from death. In this review, we summarize a handful of endogenously activated mechanisms that balance molecular cues so as to determine whether neurons recover from injury or die. We center our discussion on mechanisms that have been identified to participate in stroke, although we consider different scenarios of chronic neurodegeneration as well. We discuss two central processes that are involved in endogenous repair and that, when not regulated, could lead to tissue damage, namely, trophic support and neuroinflammation. We emphasize the need to construct integrated models of neuronal degeneration and survival that, in the end, converge in neuronal fate after injury. Under neurodegenerative conditions, endogenously activated mechanisms balance out molecular cues that determine whether neurons contend toxicity or die. Many processes involved in endogenous repair may as well lead to tissue damage depending on the strength of stimuli. Signaling mediated by trophic factors and neuroinflammation are examples of these processes as they regulate different mechanisms that mediate neuronal demise including necrosis, apoptosis, necroptosis, pyroptosis and autophagy. In this review, we discuss recent findings on balanced regulation and their involvement in neuronal death.
Collapse
Affiliation(s)
- Luis B Tovar-y-Romo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F., México
| | - Andrés Penagos-Puig
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F., México
| | - Josué O Ramírez-Jarquín
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F., México
| |
Collapse
|
16
|
Evaluating the SERCA2 and VEGF mRNAs as Potential Molecular Biomarkers of the Onset and Progression in Huntington's Disease. PLoS One 2015; 10:e0125259. [PMID: 25915065 PMCID: PMC4411078 DOI: 10.1371/journal.pone.0125259] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/20/2015] [Indexed: 01/19/2023] Open
Abstract
Abnormalities of intracellular Ca2+ homeostasis and signalling as well as the down-regulation of neurotrophic factors in several areas of the central nervous system and in peripheral tissues are hallmarks of Huntington’s disease (HD). As there is no therapy for this hereditary, neurodegenerative fatal disease, further effort should be made to slow the progression of neurodegeneration in patients through the definition of early therapeutic interventions. For this purpose, molecular biomarker(s) for monitoring disease onset and/or progression and response to treatment need to be identified. In the attempt to contribute to the research of peripheral candidate biomarkers in HD, we adopted a multiplex real-time PCR approach to analyse the mRNA level of targeted genes involved in the control of cellular calcium homeostasis and in neuroprotection. For this purpose we recruited a total of 110 subjects possessing the HD mutation at different clinical stages of the disease and 54 sex- and age-matched controls. This study provides evidence of reduced transcript levels of sarco-endoplasmic reticulum-associated ATP2A2 calcium pump (SERCA2) and vascular endothelial growth factor (VEGF) in peripheral blood mononuclear cells (PBMCs) of manifest and pre-manifest HD subjects. Our results provide a potentially new candidate molecular biomarker for monitoring the progression of this disease and contribute to understanding some early events that might have a role in triggering cellular dysfunctions in HD.
Collapse
|
17
|
O'Shea RD, Lau CL, Zulaziz N, Maclean FL, Nisbet DR, Horne MK, Beart PM. Transcriptomic analysis and 3D bioengineering of astrocytes indicate ROCK inhibition produces cytotrophic astrogliosis. Front Neurosci 2015; 9:50. [PMID: 25750613 PMCID: PMC4335181 DOI: 10.3389/fnins.2015.00050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 02/04/2015] [Indexed: 01/07/2023] Open
Abstract
Astrocytes provide trophic, structural and metabolic support to neurons, and are considered genuine targets in regenerative neurobiology, as their phenotype arbitrates brain integrity during injury. Inhibitors of Rho kinase (ROCK) cause stellation of cultured 2D astrocytes, increased L-glutamate transport, augmented G-actin, and elevated expression of BDNF and anti-oxidant genes. Here we further explored the signposts of a cytotrophic, “healthy” phenotype by data-mining of our astrocytic transcriptome in the presence of Fasudil. Gene expression profiles of motor and autophagic cellular cascades and inflammatory/angiogenic responses were all inhibited, favoring adoption of an anti-migratory phenotype. Like ROCK inhibition, tissue engineered bioscaffolds can influence the extracellular matrix. We built upon our evidence that astrocytes maintained on 3D poly-ε-caprolactone (PCL) electrospun scaffolds adopt a cytotrophic phenotype similar to that produced by Fasudil. Using these procedures, employing mature 3D cultured astrocytes, Fasudil (100 μM) or Y27632 (30 μM) added for the last 72 h of culture altered arborization, which featured numerous additional minor processes as shown by GFAP and AHNAK immunolabelling. Both ROCK inhibitors decreased F-actin, but increased G-actin labeling, indicative of disassembly of actin stress fibers. ROCK inhibitors provide additional beneficial effects for bioengineered 3D astrocytes, including enlargement of the overall arbor. Potentially, the combined strategy of bio-compatible scaffolds with ROCK inhibition offers unique advantages for the management of glial scarring. Overall these data emphasize that manipulation of the astrocyte phenotype to achieve a “healthy biology” offers new hope for the management of inflammation in neuropathologies.
Collapse
Affiliation(s)
- Ross D O'Shea
- Department of Physiology, Anatomy and Microbiology, La Trobe University Bundoora, VIC, Australia
| | - Chew L Lau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia
| | - Natasha Zulaziz
- Department of Physiology, Anatomy and Microbiology, La Trobe University Bundoora, VIC, Australia
| | - Francesca L Maclean
- Research School of Engineering, The Australian National University Canberra, ACT, Australia
| | - David R Nisbet
- Research School of Engineering, The Australian National University Canberra, ACT, Australia
| | - Malcolm K Horne
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia ; Department of Neurology, St. Vincent's Hospital Fitzroy, VIC, Australia
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
18
|
Chang KH, Wu YR, Chen YC, Chen CM. Plasma inflammatory biomarkers for Huntington's disease patients and mouse model. Brain Behav Immun 2015; 44:121-7. [PMID: 25266150 DOI: 10.1016/j.bbi.2014.09.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/19/2014] [Accepted: 09/20/2014] [Indexed: 12/19/2022] Open
Abstract
Huntington's disease (HD), caused by expanded CAG repeats encoding a polyglutamine tract in the huntingtin (HTT) protein, presents with a predominant degeneration of neurons in the striatum and cortex. Lines of evidence have observed neuroinflammation, particularly microglial activation, is involved in the pathogenesis of HD. Given that HTT is also expressed in peripheral inflammatory cells, it is possible that inflammatory changes detected in peripheral plasma may be biologically relevant and parallel the neuroinflammatory process of HD patients. By examining the expression levels of 13 microglia-derived inflammatory markers in the plasma of 5 PreHD carriers, 15 HD patients and 16 healthy controls, we found plasma levels of IL-6, MMP-9, VEGF and TGF-β1 were significantly increased in HD patients when compared with the controls, while plasma level of IL-18 were significantly reduced in HD patients compared with controls. Plasma level of IL-6 was reversely correlated with the UHDRS independence scale and functional capacity. To understand the temporal correlation between these inflammatory markers and HD progression, their levels were further tested in plasma from R6/2 mouse HD model at different ages. In rotarod test, R6/2 HD mice started to manifest HD phenotype at 7.5 weeks of age. Higher plasma VEGF levels of R6/2 mice than those of age-matched wild-type (WT) littermates were noted from 7 (presymptomatic stage) to 13 weeks of age (late symptomatic stage). The plasma IL-6 levels of R6/2 mice were higher than those of the WT littermates from 9 (early symptomatic stage) to 13 weeks of age. R6/2 mice demonstrated higher MMP-9 and TGF-β1 levels than their WT littermates from 11 (middle symptomatic stage) to 13 weeks of age. In contrast, the plasma IL-18 level was lower than those in WT littermates since 11 weeks of age. These altered expressions of inflammatory markers may serve as the potential biomarkers for HD onset and progression. Specific inhibition/activation of these inflammatory markers may be the targets of HD drug development.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Chun Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
19
|
Agustín-Pavón C, Isalan M. Synthetic biology and therapeutic strategies for the degenerating brain: Synthetic biology approaches can transform classical cell and gene therapies, to provide new cures for neurodegenerative diseases. Bioessays 2014; 36:979-90. [PMID: 25100403 PMCID: PMC4312882 DOI: 10.1002/bies.201400094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Synthetic biology is an emerging engineering discipline that attempts to design and rewire biological components, so as to achieve new functions in a robust and predictable manner. The new tools and strategies provided by synthetic biology have the potential to improve therapeutics for neurodegenerative diseases. In particular, synthetic biology will help design small molecules, proteins, gene networks, and vectors to target disease-related genes. Ultimately, new intelligent delivery systems will provide targeted and sustained therapeutic benefits. New treatments will arise from combining ‘protect and repair’ strategies: the use of drug treatments, the promotion of neurotrophic factor synthesis, and gene targeting. Going beyond RNAi and artificial transcription factors, site-specific genome modification is likely to play an increasing role, especially with newly available gene editing tools such as CRISPR/Cas9 systems. Taken together, these advances will help develop safe and long-term therapies for many brain diseases in human patients.
Collapse
|