1
|
Ren Z, Mao X, Zhang Z, Wang W. The impact of sleep deprivation on cognitive function in healthy adults: insights from auditory P300 and reaction time analysis. Front Neurosci 2025; 19:1559969. [PMID: 40270765 PMCID: PMC12014645 DOI: 10.3389/fnins.2025.1559969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
Objective This study aims to explore the effects of sleep deprivation on cognitive function in healthy adults, using auditory P300 event-related potentials and subjective reaction time as key assessment metrics. Methods High-density electroencephalography (EEG) and the oddball paradigm were utilized to collect P300 event-related potentials (ERPs) before and after the sleep deprivation intervention, with a record of subjective reaction time. Participants were classified into acute sleep deprivation group and chronic sleep deprivation group based on the duration of their night shift history. Results Following sleep deprivation, a significant prolongation in P300 latency and reaction time was observed among 26 subjects (P < 0.05). Specifically, the reaction time in the acute group increased significantly by 83.69 ms after sleep deprivation (P < 0.05). In contrast, the reaction time in the chronic group exhibited only a minor increase of 6.54 ms (P < 0.05). Furthermore, a significant interaction effect between the duration of night shift history and sleep condition on reaction time was identified [F (aXb) = 4.736, P = 0.040, η p 2 = 0.165], suggesting that the influence of sleep deprivation on reaction time varies between the chronic and acute groups. Conclusion Sleep deprivation induces cognitive impairment, with the acute sleep deprivation group experiencing more severe deficits. In contrast, the chronic sleep deprivation group demonstrated milder but chronic cognitive impairment.
Collapse
Affiliation(s)
- Zhongkai Ren
- Department of Otorhinolaryngology-Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China
- Institute of Otolaryngology of Tianjin, Tianjin, China
- Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China
- Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China
- Otolaryngology Clinical Quality Control Centre, Tianjin, China
- First Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Xiang Mao
- Department of Otorhinolaryngology-Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China
- Institute of Otolaryngology of Tianjin, Tianjin, China
- Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China
- Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China
- Otolaryngology Clinical Quality Control Centre, Tianjin, China
| | - Ziyue Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China
- Institute of Otolaryngology of Tianjin, Tianjin, China
- Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China
- Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China
- Otolaryngology Clinical Quality Control Centre, Tianjin, China
| | - Wei Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Tianjin First Central Hospital, Tianjin, China
- Institute of Otolaryngology of Tianjin, Tianjin, China
- Key Laboratory of Auditory Speech and Balance Medicine, Tianjin, China
- Key Medical Discipline of Tianjin (Otolaryngology), Tianjin, China
- Otolaryngology Clinical Quality Control Centre, Tianjin, China
| |
Collapse
|
2
|
Butt TH, Tobiume M, Re DB, Kariya S. Physical Exercise Counteracts Aging-Associated White Matter Demyelination Causing Cognitive Decline. Aging Dis 2024; 15:2136-2148. [PMID: 38377028 PMCID: PMC11346408 DOI: 10.14336/ad.2024.0216] [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: 01/14/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
In the central nervous system, oligodendrocytes wrap around neuronal axons to form myelin, an insulating layer or sheath that allows for the efficient conductance of action potentials. In addition to structural insulation, myelin provides encased axons with nutrient, metabolic and defensive support. Demyelination, or myelin loss, can therefore cause axonal dysfunction, leading to neurological impairment and disease. In Alzheimer's disease (AD), progressive white matter demyelination is acknowledged as one of the earliest pathologies preceding symptom onset. Unfortunately, current pharmacotherapy for slowing demyelination or promoting remyelination in AD is nonexistent. Exercise is recognized for its wide-ranging benefits to human health, including improved mental health and the prevention of lifestyle-related diseases. Mounting evidence suggests the contribution of physical activity in delaying the progression of dementia in elderly populations. Recent mechanistic studies have shown that exercise facilitates myelination in the brain through the vitalization of intrinsic pro-myelination cues, such as increased neurotrophic factors and electrical activity. In this review, we summarize and discuss the potential of physical exercise on counteracting aging-associated white matter demyelination, which causes cognitive decline in AD. We highlight the need of further basic and clinical research investigations on this topic to establish novel approaches for healthy and improved brain aging.
Collapse
Affiliation(s)
- Tanya H Butt
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Makoto Tobiume
- Unit for Respiratory System & Dementia in the Division of Internal Medicine, Katsuren Hospital, Itoman, Okinawa, Japan
| | - Diane B Re
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
- NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
| | - Shingo Kariya
- Unit for Nervous System & Dementia in the Division of Internal Medicine, Katsuren Hospital, Itoman, Okinawa, Japan
| |
Collapse
|
3
|
D'Egidio F, Castelli V, Lombardozzi G, Ammannito F, Cimini A, d'Angelo M. Therapeutic advances in neural regeneration for Huntington's disease. Neural Regen Res 2024; 19:1991-1997. [PMID: 38227527 DOI: 10.4103/1673-5374.390969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/03/2023] [Indexed: 01/17/2024] Open
Abstract
Huntington's disease is a neurodegenerative disease caused by the expansion mutation of a cytosine-adenine-guanine triplet in the exon 1 of the HTT gene which is responsible for the production of the huntingtin (Htt) protein. In physiological conditions, Htt is involved in many cellular processes such as cell signaling, transcriptional regulation, energy metabolism regulation, DNA maintenance, axonal trafficking, and antiapoptotic activity. When the genetic alteration is present, the production of a mutant version of Htt (mHtt) occurs, which is characterized by a plethora of pathogenic activities that, finally, lead to cell death. Among all the cells in which mHtt exerts its dangerous activity, the GABAergic Medium Spiny Neurons seem to be the most affected by the mHtt-induced excitotoxicity both in the cortex and in the striatum. However, as the neurodegeneration proceeds ahead the neuronal loss grows also in other brain areas such as the cerebellum, hypothalamus, thalamus, subthalamic nucleus, globus pallidus, and substantia nigra, determining the variety of symptoms that characterize Huntington's disease. From a clinical point of view, Huntington's disease is characterized by a wide spectrum of symptoms spanning from motor impairment to cognitive disorders and dementia. Huntington's disease shows a prevalence of around 3.92 cases every 100,000 worldwide and an incidence of 0.48 new cases every 100,000/year. To date, there is no available cure for Huntington's disease. Several treatments have been developed so far, aiming to reduce the severity of one or more symptoms to slow down the inexorable decline caused by the disease. In this context, the search for reliable strategies to target the different aspects of Huntington's disease become of the utmost interest. In recent years, a variety of studies demonstrated the detrimental role of neuronal loss in Huntington's disease condition highlighting how the replacement of lost cells would be a reasonable strategy to overcome the neurodegeneration. In this view, numerous have been the attempts in several preclinical models of Huntington's disease to evaluate the feasibility of invasive and non-invasive approaches. Thus, the aim of this review is to offer an overview of the most appealing approaches spanning from stem cell-based cell therapy to extracellular vesicles such as exosomes in light of promoting neurogenesis, discussing the results obtained so far, their limits and the future perspectives regarding the neural regeneration in the context of Huntington's disease.
Collapse
Affiliation(s)
- Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | |
Collapse
|
4
|
Andreadou M, Ingelfinger F, De Feo D, Cramer TLM, Tuzlak S, Friebel E, Schreiner B, Eede P, Schneeberger S, Geesdorf M, Ridder F, Welsh CA, Power L, Kirschenbaum D, Tyagarajan SK, Greter M, Heppner FL, Mundt S, Becher B. IL-12 sensing in neurons induces neuroprotective CNS tissue adaptation and attenuates neuroinflammation in mice. Nat Neurosci 2023; 26:1701-1712. [PMID: 37749256 PMCID: PMC10545539 DOI: 10.1038/s41593-023-01435-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 08/15/2023] [Indexed: 09/27/2023]
Abstract
Interleukin-12 (IL-12) is a potent driver of type 1 immunity. Paradoxically, in autoimmune conditions, including of the CNS, IL-12 reduces inflammation. The underlying mechanism behind these opposing properties and the involved cellular players remain elusive. Here we map IL-12 receptor (IL-12R) expression to NK and T cells as well as neurons and oligodendrocytes. Conditionally ablating the IL-12R across these cell types in adult mice and assessing their susceptibility to experimental autoimmune encephalomyelitis revealed that the neuroprotective role of IL-12 is mediated by neuroectoderm-derived cells, specifically neurons, and not immune cells. In human brain tissue from donors with multiple sclerosis, we observe an IL-12R distribution comparable to mice, suggesting similar mechanisms in mice and humans. Combining flow cytometry, bulk and single-nucleus RNA sequencing, we reveal an IL-12-induced neuroprotective tissue adaption preventing early neurodegeneration and sustaining trophic factor release during neuroinflammation, thereby maintaining CNS integrity in mice.
Collapse
Affiliation(s)
- Myrto Andreadou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Teresa L M Cramer
- Institute of Pharmacology and Toxicology, Neurodevelopmental Pharmacology, University of Zurich, Zurich, Switzerland
| | - Selma Tuzlak
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ekaterina Friebel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Schreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Pascale Eede
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Shirin Schneeberger
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Maria Geesdorf
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederike Ridder
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Christina A Welsh
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Laura Power
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Daniel Kirschenbaum
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Systems Immunology, Weizmann Institute, Rehovot, Israel
| | - Shiva K Tyagarajan
- Institute of Pharmacology and Toxicology, Neurodevelopmental Pharmacology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Frank L Heppner
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Thümmler K, Wrzos C, Franz J, McElroy D, Cole JJ, Hayden L, Arseni D, Schwarz F, Junker A, Edgar JM, Kügler S, Neef A, Wolf F, Stadelmann C, Linington C. Fibroblast growth factor 9 (FGF9)-mediated neurodegeneration: Implications for progressive multiple sclerosis? Neuropathol Appl Neurobiol 2023; 49:e12935. [PMID: 37705188 DOI: 10.1111/nan.12935] [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: 02/12/2023] [Revised: 08/22/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
AIMS Fibroblast growth factor (FGF) signalling is dysregulated in multiple sclerosis (MS) and other neurological and psychiatric conditions, but there is little or no consensus as to how individual FGF family members contribute to disease pathogenesis. Lesion development in MS is associated with increased expression of FGF1, FGF2 and FGF9, all of which modulate remyelination in a variety of experimental settings. However, FGF9 is also selectively upregulated in major depressive disorder (MDD), prompting us to speculate it may also have a direct effect on neuronal function and survival. METHODS Transcriptional profiling of myelinating cultures treated with FGF1, FGF2 or FGF9 was performed, and the effects of FGF9 on cortical neurons investigated using a combination of transcriptional, electrophysiological and immunofluorescence microscopic techniques. The in vivo effects of FGF9 were explored by stereotactic injection of adeno-associated viral (AAV) vectors encoding either FGF9 or EGFP into the rat motor cortex. RESULTS Transcriptional profiling of myelinating cultures after FGF9 treatment revealed a distinct neuronal response with a pronounced downregulation of gene networks associated with axonal transport and synaptic function. In cortical neuronal cultures, FGF9 also rapidly downregulated expression of genes associated with synaptic function. This was associated with a complete block in the development of photo-inducible spiking activity, as demonstrated using multi-electrode recordings of channel rhodopsin-transfected rat cortical neurons in vitro and, ultimately, neuronal cell death. Overexpression of FGF9 in vivo resulted in rapid loss of neurons and subsequent development of chronic grey matter lesions with neuroaxonal reduction and ensuing myelin loss. CONCLUSIONS These observations identify overexpression of FGF9 as a mechanism by which neuroaxonal pathology could develop independently of immune-mediated demyelination in MS. We suggest targeting neuronal FGF9-dependent pathways may provide a novel strategy to slow if not halt neuroaxonal atrophy and loss in MS, MDD and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Katja Thümmler
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Claudia Wrzos
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Jonas Franz
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Göttingen Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
| | - Daniel McElroy
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - John J Cole
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Lorna Hayden
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Diana Arseni
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Friedrich Schwarz
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Junker
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Department of Neuropathology, University Hospital Essen, Essen, Germany
| | - Julia M Edgar
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Sebastian Kügler
- Institute for Neurology, University Medical Center Göttingen, Göttingen, Germany
- Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Andreas Neef
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Göttingen Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Fred Wolf
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Göttingen Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
- Cluster of Excellence Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells (MBExC), University of Goettingen, Göttingen, Germany
| | - Christine Stadelmann
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells (MBExC), University of Goettingen, Göttingen, Germany
| | | |
Collapse
|
6
|
Zhai W, Zhang T, Jin Y, Huang S, Xu M, Pan J. The fibroblast growth factor system in cognitive disorders and dementia. Front Neurosci 2023; 17:1136266. [PMID: 37214403 PMCID: PMC10196031 DOI: 10.3389/fnins.2023.1136266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Cognitive impairment is the core precursor to dementia and other cognitive disorders. Current hypotheses suggest that they share a common pathological basis, such as inflammation, restricted neurogenesis, neuroendocrine disorders, and the destruction of neurovascular units. Fibroblast growth factors (FGFs) are cell growth factors that play essential roles in various pathophysiological processes via paracrine or autocrine pathways. This system consists of FGFs and their receptors (FGFRs), which may hold tremendous potential to become a new biological marker in the diagnosis of dementia and other cognitive disorders, and serve as a potential target for drug development against dementia and cognitive function impairment. Here, we review the available evidence detailing the relevant pathways mediated by multiple FGFs and FGFRs, and recent studies examining their role in the pathogenesis and treatment of cognitive disorders and dementia.
Collapse
|
7
|
Jang W, Mun SJ, Kim SY, Bong KW. Controlled growth factor delivery via a degradable poly(lactic acid) hydrogel microcarrier synthesized using degassed micromolding lithography. Colloids Surf B Biointerfaces 2023; 222:113088. [PMID: 36577342 DOI: 10.1016/j.colsurfb.2022.113088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Controlled and targeted delivery of growth factors to biological environments is important for tissue regeneration. Polylactic acid (PLA) hydrogel microparticles are attractive carriers for the delivery of therapeutic cargoes based on their superior biocompatibility and biodegradability, uniform encapsulation of cargoes, and non-requirement of organic solvents during particle synthesis. In this study, we newly present controlled growth factor delivery utilizing PLA-based hydrogel microcarriers synthesized via degassed micromolding lithography (DML). Based on the direct gelation procedure from the single-phase aqueous precursor in DML, bovine serum albumin, a model protein of growth factor, and fibroblast growth factor were encapsulated into microparticles with uniform distribution. In addition, by tuning the monomer concentration and adding a hydrolytically stable crosslinker, the release of encapsulated cargoes was efficiently controlled and extended to 2 weeks. Finally, we demonstrated the biological activity of encapsulated FGF-2 in PLA-based microparticles using a fibroblast proliferation assay.
Collapse
Affiliation(s)
- Wookyoung Jang
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Seok Joon Mun
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Soung-Yon Kim
- Department of Orthopaedic Surgery, Kangwon National University Hospital, Baengnyeong-ro 156, Chuncheon-si, Gangwon-do 24289, Republic of Korea.
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.
| |
Collapse
|
8
|
Boado RJ. IgG Fusion Proteins for Brain Delivery of Biologics via Blood-Brain Barrier Receptor-Mediated Transport. Pharmaceutics 2022; 14:pharmaceutics14071476. [PMID: 35890374 PMCID: PMC9322584 DOI: 10.3390/pharmaceutics14071476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 01/01/2023] Open
Abstract
The treatment of neurological disorders with large-molecule biotherapeutics requires that the therapeutic drug be transported across the blood–brain barrier (BBB). However, recombinant biotherapeutics, such as neurotrophins, enzymes, decoy receptors, and monoclonal antibodies (MAb), do not cross the BBB. These biotherapeutics can be re-engineered as brain-penetrating bifunctional IgG fusion proteins. These recombinant proteins comprise two domains, the transport domain and the therapeutic domain, respectively. The transport domain is an MAb that acts as a molecular Trojan horse by targeting a BBB-specific endogenous receptor that induces receptor-mediated transcytosis into the brain, such as the human insulin receptor (HIR) or the transferrin receptor (TfR). The therapeutic domain of the IgG fusion protein exerts its pharmacological effect in the brain once across the BBB. A generation of bifunctional IgG fusion proteins has been engineered using genetically engineered MAbs directed to either the BBB HIR or TfR as the transport domain. These IgG fusion proteins were validated in animal models of lysosomal storage disorders; acute brain conditions, such as stroke; or chronic neurodegeneration, such as Parkinson’s disease and Alzheimer’s disease. Human phase I–III clinical trials were also completed for Hurler MPSI and Hunter MPSII using brain-penetrating IgG-iduronidase and -iduronate-2-sulfatase fusion protein, respectively.
Collapse
Affiliation(s)
- Ruben J Boado
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
Li H, Gan X, Pan L, Zhang Y, Hu X, Wang Z. EGF/bFGF promotes survival, migration and differentiation into neurons of GFP-labeled rhesus monkey neural stem cells xenografted into the rat brain. Biochem Biophys Res Commun 2022; 620:76-82. [DOI: 10.1016/j.bbrc.2022.06.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/13/2022] [Accepted: 06/23/2022] [Indexed: 11/02/2022]
|
10
|
Zhao Y, Wang Q, Xie C, Cai Y, Chen X, Hou Y, He L, Li J, Yao M, Chen S, Wu W, Chen X, Hong A. Peptide ligands targeting FGF receptors promote recovery from dorsal root crush injury via AKT/mTOR signaling. Am J Cancer Res 2021; 11:10125-10147. [PMID: 34815808 PMCID: PMC8581430 DOI: 10.7150/thno.62525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Fibroblast growth factor receptors (FGFRs) are key targets for nerve regeneration and repair. The therapeutic effect of exogenous recombinant FGFs in vivo is limited due to their high molecular weight. Small peptides with low molecular weight, easy diffusion, low immunogenicity, and nontoxic metabolite formation are potential candidates. The present study aimed to develop a novel low-molecular-weight peptide agonist of FGFR to promote nerve injury repair. Methods: Phage display technology was employed to screen peptide ligands targeting FGFR2. The peptide ligand affinity for FGFRs was detected by isothermal titration calorimetry. Structural biology-based computer virtual analysis was used to characterize the interaction between the peptide ligand and FGFR2. The peptide ligand effect on axon growth, regeneration, and behavioral recovery of sensory neurons was determined in the primary culture of sensory neurons and dorsal root ganglia (DRG) explants in vitro and a rat spinal dorsal root injury (DRI) model in vivo. The peptide ligand binding to other membrane receptors was characterized by surface plasmon resonance (SPR) and liquid chromatography-mass spectrometry (LC-MS)/MS. Intracellular signaling pathways primarily affected by the peptide ligand were characterized by phosphoproteomics, and related pathways were verified using specific inhibitors. Results: We identified a novel FGFR-targeting small peptide, CH02, with seven amino acid residues. CH02 activated FGFR signaling through high-affinity binding with the extracellular segment of FGFRs and also had an affinity for several receptor tyrosine kinase (RTK) family members, including VEGFR2. In sensory neurons cultured in vitro, CH02 maintained the survival of neurons and promoted axon growth. Simultaneously, CH02 robustly enhanced nerve regeneration and sensory-motor behavioral recovery after DRI in rats. CH02-induced activation of FGFR signaling promoted nerve regeneration primarily via AKT and ERK signaling downstream of FGFRs. Activation of mTOR downstream of AKT signaling augmented axon growth potential in response to CH02. Conclusion: Our study revealed the significant therapeutic effect of CH02 on strengthening nerve regeneration and suggested a strategy for treating peripheral and central nervous system injuries.
Collapse
|
11
|
Gatto RG, Weissmann C. Preliminary examination of early neuroconnectivity features in the R6/1 mouse model of Huntington's disease by ultra-high field diffusion MRI. Neural Regen Res 2021; 17:983-986. [PMID: 34558512 PMCID: PMC8552860 DOI: 10.4103/1673-5374.324831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
During the last decades, advances in the understanding of genetic, cellular, and microstructural alterations associated to Huntington's disease (HD) have improved the understanding of this progressive and fatal illness. However, events related to early neuropathological events, neuroinflammation, deterioration of neuronal connectivity and compensatory mechanisms still remain vastly unknown. Ultra-high field diffusion MRI (UHFD-MRI) techniques can contribute to a more comprehensive analysis of the early microstructural changes observed in HD. In addition, it is possible to evaluate if early imaging microstructural parameters might be linked to histological biomarkers. Moreover, qualitative studies analyzing histological complexity in brain areas susceptible to neurodegeneration could provide information on inflammatory events, compensatory increase of neuroconnectivity and mechanisms of brain repair and regeneration. The application of ultra-high field diffusion-MRI technology in animal models, particularly the R6/1 mice (a common preclinical mammalian model of HD), provide the opportunity to analyze alterations in a physiologically intact model of the disease. Although some disparities in volumetric changes across different brain structures between preclinical and clinical models has been documented, further application of different diffusion MRI techniques used in combination like diffusion tensor imaging, and neurite orientation dispersion and density imaging have proved effective in characterizing early parameters associated to alteration in water diffusion exchange within intracellular and extracellular compartments in brain white and grey matter. Thus, the combination of diffusion MRI imaging techniques and more complex neuropathological analysis could accelerate the discovery of new imaging biomarkers and the early diagnosis and neuromonitoring of patients affected with HD.
Collapse
Affiliation(s)
- Rodolfo G Gatto
- Department of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA
| | - Carina Weissmann
- Instituto de Fisiología Biología Molecular y Neurociencias-IFIBYNE-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
12
|
Dias MH, Bernards R. Playing cancer at its own game: activating mitogenic signaling as a paradoxical intervention. Mol Oncol 2021; 15:1975-1985. [PMID: 33955157 PMCID: PMC8333773 DOI: 10.1002/1878-0261.12979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/12/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
In psychotherapy, paradoxical interventions are characterized by a deliberate reinforcement of the pathological behavior to improve the clinical condition. Such a counter-intuitive approach can be considered when more conventional interventions fail. The development of targeted cancer therapies has enabled the selective inhibition of activated oncogenic signaling pathways. However, in advanced cancers, such therapies, on average, deliver modest benefits due to the development of resistance. Here, we review the perspective of a 'paradoxical intervention' in cancer therapy: rather than attempting to inhibit oncogenic signaling, the proposed therapy would further activate mitogenic signaling to disrupt the labile homeostasis of cancer cells and overload stress response pathways. Such overactivation can potentially be combined with stress-targeted drugs to kill overstressed cancer cells. Although counter-intuitive, such an approach exploits intrinsic and ubiquitous differences between normal and cancer cells. We discuss the background underlying this unconventional approach and how such intervention might address some current challenges in cancer therapy.
Collapse
Affiliation(s)
- Matheus Henrique Dias
- Division of Molecular CarcinogenesisOncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - René Bernards
- Division of Molecular CarcinogenesisOncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
13
|
Lee B, Shin M, Park Y, Won SY, Cho KS. Physical Exercise-Induced Myokines in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22115795. [PMID: 34071457 PMCID: PMC8198301 DOI: 10.3390/ijms22115795] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS), are disorders characterized by progressive degeneration of the nervous system. Currently, there is no disease-modifying treatments for most NDs. Meanwhile, numerous studies conducted on human and animal models over the past decades have showed that exercises had beneficial effects on NDs. Inter-tissue communication by myokine, a peptide produced and secreted by skeletal muscles during exercise, is thought to be an important underlying mechanism for the advantages. Here, we reviewed studies about the effects of myokines regulated by exercise on NDs and their mechanisms. Myokines could exert beneficial effects on NDs through a variety of regulatory mechanisms, including cell survival, neurogenesis, neuroinflammation, proteostasis, oxidative stress, and protein modification. Studies on exercise-induced myokines are expected to provide a novel strategy for treating NDs, for which there are no adequate treatments nowadays. To date, only a few myokines have been investigated for their effects on NDs and studies on mechanisms involved in them are in their infancy. Therefore, future studies are needed to discover more myokines and test their effects on NDs.
Collapse
Affiliation(s)
- Banseok Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - Myeongcheol Shin
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - Youngjae Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - So-Yoon Won
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.-Y.W.); (K.S.C.); Tel.: +82-10-3688-5474 (S.-Y.W.); Tel.: +82-2-450-3424 (K.S.C.)
| | - Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.-Y.W.); (K.S.C.); Tel.: +82-10-3688-5474 (S.-Y.W.); Tel.: +82-2-450-3424 (K.S.C.)
| |
Collapse
|
14
|
Clozapine protects adult neural stem cells from ketamine-induced cell death in correlation with decreased apoptosis and autophagy. Biosci Rep 2021; 40:221825. [PMID: 31919522 PMCID: PMC6981094 DOI: 10.1042/bsr20193156] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
Adult neurogenesis, the production of newborn neurons from neural stem cells (NSCs) has been suggested to be decreased in patients with schizophrenia. A similar finding was observed in an animal model of schizophrenia, as indicated by decreased bromodeoxyuridine (BrdU) labelling cells in response to a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist. The antipsychotic drug clozapine was shown to counteract the observed decrease in BrdU-labelled cells in hippocampal dentate gyrus (DG). However, phenotypic determination by immunohistochemistry analysis could not reveal whether BrdU-positive cells were indeed NSCs. Using a previously established cell model for analysing NSC protection in vitro, we investigated a protective effect of clozapine on NSCs. Primary NSCs were isolated from the mouse subventricular zone (SVZ), we show that clozapine had a NSC protective activity alone, as evident by employing an ATP cell viability assay. In contrast, haloperidol did not show any NSC protective properties. Subsequently, cells were exposed to the non-competitive NMDA-receptor antagonist ketamine. Clozapine, but not haloperidol, had a NSC protective/anti-apoptotic activity against ketamine-induced cytotoxicity. The observed NSC protective activity of clozapine was associated with increased expression of the anti-apoptotic marker Bcl-2, decreased expression of the pro-apoptotic cleaved form of caspase-3 and associated with decreased expression of the autophagosome marker 1A/1B-light chain 3 (LC3-II). Collectively, our findings suggest that clozapine may have a protective/anti-apoptotic effect on NSCs, supporting previous in vivo observations, indicating a neurogenesis-promoting activity for clozapine. If the data are further confirmed in vivo, the results may encourage an expanded use of clozapine to restore impaired neurogenesis in schizophrenia.
Collapse
|
15
|
Yusuf IO, Chen HM, Cheng PH, Chang CY, Tsai SJ, Chuang JI, Wu CC, Huang BM, Sun HS, Chen CM, Yang SH. Fibroblast Growth Factor 9 Stimulates Neuronal Length Through NF-kB Signaling in Striatal Cell Huntington's Disease Models. Mol Neurobiol 2021; 58:2396-2406. [PMID: 33421017 DOI: 10.1007/s12035-020-02220-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/18/2020] [Indexed: 12/29/2022]
Abstract
Proper development of neuronal cells is important for brain functions, and impairment of neuronal development may lead to neuronal disorders, implying that improvement in neuronal development may be a therapeutic direction for these diseases. Huntington's disease (HD) is a neurodegenerative disease characterized by impairment of neuronal structures, ultimately leading to neuronal death and dysfunctions of the central nervous system. Based on previous studies, fibroblast growth factor 9 (FGF9) may provide neuroprotective functions in HD, and FGFs may enhance neuronal development and neurite outgrowth. However, whether FGF9 can provide neuronal protective functions through improvement of neuronal morphology in HD is still unclear. Here, we study the effects of FGF9 on neuronal length in HD and attempt to understand the related working mechanisms. Taking advantage of striatal cell lines from HD knock-in mice, we found that FGF9 increases total neuronal length and upregulates several structural and synaptic proteins under HD conditions. In addition, activation of nuclear factor kappa B (NF-kB) signaling by FGF9 was observed to be significant in HD cells, and blockage of NF-kB leads to suppression of these structural and synaptic proteins induced by FGF9, suggesting the involvement of NF-kB signaling in these effects of FGF9. Taken these results together, FGF9 may enhance total neuronal length through upregulation of NF-kB signaling, and this mechanism could serve as an important mechanism for neuroprotective functions of FGF9 in HD.
Collapse
Affiliation(s)
- Issa Olakunle Yusuf
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, 11529, Taiwan.,Institute of Clinical Medicine, Taipei, Taiwan.,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hsiu-Mei Chen
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Pei-Hsun Cheng
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chih-Yi Chang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Shaw-Jenq Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Institute of Basic Medical Sciences, Taipei, Taiwan
| | - Jih-Ing Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Institute of Basic Medical Sciences, Taipei, Taiwan
| | - Chia-Ching Wu
- Institute of Basic Medical Sciences, Taipei, Taiwan.,Department of Cell Biology and Anatomy, Taipei, Taiwan
| | - Bu-Miin Huang
- Institute of Basic Medical Sciences, Taipei, Taiwan.,Department of Cell Biology and Anatomy, Taipei, Taiwan
| | - H Sunny Sun
- Institute of Basic Medical Sciences, Taipei, Taiwan.,Institute of Molecular Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Shang-Hsun Yang
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, 11529, Taiwan. .,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan. .,Institute of Basic Medical Sciences, Taipei, Taiwan.
| |
Collapse
|
16
|
Jurkowski MP, Bettio L, K. Woo E, Patten A, Yau SY, Gil-Mohapel J. Beyond the Hippocampus and the SVZ: Adult Neurogenesis Throughout the Brain. Front Cell Neurosci 2020; 14:576444. [PMID: 33132848 PMCID: PMC7550688 DOI: 10.3389/fncel.2020.576444] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Convincing evidence has repeatedly shown that new neurons are produced in the mammalian brain into adulthood. Adult neurogenesis has been best described in the hippocampus and the subventricular zone (SVZ), in which a series of distinct stages of neuronal development has been well characterized. However, more recently, new neurons have also been found in other brain regions of the adult mammalian brain, including the hypothalamus, striatum, substantia nigra, cortex, and amygdala. While some studies have suggested that these new neurons originate from endogenous stem cell pools located within these brain regions, others have shown the migration of neurons from the SVZ to these regions. Notably, it has been shown that the generation of new neurons in these brain regions is impacted by neurologic processes such as stroke/ischemia and neurodegenerative disorders. Furthermore, numerous factors such as neurotrophic support, pharmacologic interventions, environmental exposures, and stem cell therapy can modulate this endogenous process. While the presence and significance of adult neurogenesis in the human brain (and particularly outside of the classical neurogenic regions) is still an area of debate, this intrinsic neurogenic potential and its possible regulation through therapeutic measures present an exciting alternative for the treatment of several neurologic conditions. This review summarizes evidence in support of the classic and novel neurogenic zones present within the mammalian brain and discusses the functional significance of these new neurons as well as the factors that regulate their production. Finally, it also discusses the potential clinical applications of promoting neurogenesis outside of the classical neurogenic niches, particularly in the hypothalamus, cortex, striatum, substantia nigra, and amygdala.
Collapse
Affiliation(s)
- Michal P. Jurkowski
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Luis Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Emma K. Woo
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Anna Patten
- Centre for Interprofessional Clinical Simulation Learning (CICSL), Royal Jubilee Hospital, Victoria, BC, Canada
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Joana Gil-Mohapel
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
17
|
Sénécal V, Barat C, Tremblay MJ. The delicate balance between neurotoxicity and neuroprotection in the context of HIV-1 infection. Glia 2020; 69:255-280. [PMID: 32910482 DOI: 10.1002/glia.23904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/17/2022]
Abstract
Human immunodeficiency virus type-1 (HIV-1) causes a spectrum of neurological impairments, termed HIV-associated neurocognitive disorder (HAND), following the infiltration of infected cells into the brain. Even though the implementation of antiretroviral therapy reduced the systemic viral load, the prevalence of HAND remains unchanged and infected patients develop persisting neurological disturbances affecting their quality of life. As a result, HAND have gained importance in basic and clinical researches, warranting the need of developing new adjunctive treatments. Nonetheless, a better understanding of the molecular and cellular mechanisms remains necessary. Several studies consolidated their efforts into elucidating the neurotoxic signaling leading to HAND including the deleterious actions of HIV-1 viral proteins and inflammatory mediators. However, the scope of these studies is not sufficient to address all the complexity related to HAND development. Fewer studies focused on an altered neuroprotective capacity of the brain to respond to HIV-1 infection. Neurotrophic factors are endogenous polyproteins involved in neuronal survival, synaptic plasticity, and neurogenesis. Any defects in the processing or production of these crucial factors might compose a risk factor rendering the brain more vulnerable to neuronal damages. Due to their essential roles, they have been investigated for their diverse interplays with HIV-1 infection. In this review, we present a complete description of the neurotrophic factors involved in HAND. We discuss emerging concepts for their therapeutic applications and summarize the complex mechanisms that down-regulate their production in favor of a neurotoxic environment. For certain factors, we finally address opposing roles that rather lead to increased inflammation.
Collapse
Affiliation(s)
- Vincent Sénécal
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec, Quebec, Canada
| | - Corinne Barat
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec, Quebec, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec, Quebec, Canada.,Département de Microbiologie-infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
18
|
Hayashida M, Hashioka S, Hayashida K, Miura S, Tsuchie K, Araki T, Izuhara M, Kanayama M, Otsuki K, Nagahama M, Jaya MA, Arauchi R, Wake R, Oh-Nishi A, Horiguchi J, Miyaoka T, Inagaki M, Morita E. Low Serum Levels of Fibroblast Growth Factor 2 in Gunn Rats: A Hyperbilirubinemia Animal Model of Schizophrenic Symptoms. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:503-508. [PMID: 32729434 DOI: 10.2174/1871527319999200729153907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/06/2020] [Accepted: 06/07/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Fibroblast Growth Factor (FGF) 2 (also referred to as basic FGF) is a multifunctional growth factor that plays a pivotal role in the pro-survival, pro-migration and prodifferentiation of neurons. METHOD Because alterations in FGF2 levels are suggested to contribute to the pathogenesis of schizophrenia, we investigated serum levels of FGF2 in the Gunn rat, a hyperbilirubinemia animal model of schizophrenic symptoms. RESULTS The enzyme-linked immunosorbent assay showed that the serum levels of FGF2 in Gunn rats were 5.09 ± 0.236 pg/mL, while those in the normal strain Wistar rats, serum levels were 11.90 ± 2.142 pg/mL. The serum FGF2 levels in Gunn rats were significantly lower than those in Wistar rats. We also measured serum levels of Unconjugated Bilirubin (UCB) and found a significant negative correlation between UCB and FGF2 in terms of serum levels in all the rats studied. CONCLUSION Since it is known that FGF2 regulates dopaminergic neurons and have antineuroinflammatory effects, our finding suggests that low FGF2 levels may contribute to the pathogenesis of schizophrenia, in which imbalanced dopamin-ergic signaling and neuroinflammation are supposed to play certain roles.
Collapse
Affiliation(s)
- Maiko Hayashida
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Sadayuki Hashioka
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Kenji Hayashida
- Division of Plastic Surgery, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Shoko Miura
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Keiko Tsuchie
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Tomoko Araki
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Muneto Izuhara
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Misako Kanayama
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Koji Otsuki
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Michiharu Nagahama
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Muhammad Alim Jaya
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Ryosuke Arauchi
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Rei Wake
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Arata Oh-Nishi
- Department of Immuno-Neuropsychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Jun Horiguchi
- Department of Immuno-Neuropsychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Tsuyoshi Miyaoka
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Masatoshi Inagaki
- Department of Psychiatry, Faculty of Medicine, Shimane University, Matsue, Japan
| | - Eishin Morita
- Department of Dermatology, Faculty of Medicine, Shimane University, Matsue, Japan
| |
Collapse
|
19
|
Segarra M, Aburto MR, Hefendehl J, Acker-Palmer A. Neurovascular Interactions in the Nervous System. Annu Rev Cell Dev Biol 2020; 35:615-635. [PMID: 31590587 DOI: 10.1146/annurev-cellbio-100818-125142] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular cross talk between the nervous and vascular systems is necessary to maintain the correct coupling of organ structure and function. Molecular pathways shared by both systems are emerging as major players in the communication of the neuronal compartment with the endothelium. Here we review different aspects of this cross talk and how vessels influence the development and homeostasis of the nervous system. Beyond the classical role of the vasculature as a conduit to deliver oxygen and metabolites needed for the energy-demanding neuronal compartment, vessels emerge as powerful signaling systems that control and instruct a variety of cellular processes during the development of neurons and glia, such as migration, differentiation, and structural connectivity. Moreover, a broad spectrum of mild to severe vascular dysfunctions occur in various pathologies of the nervous system, suggesting that mild structural and functional changes at the neurovascular interface may underlie cognitive decline in many of these pathological conditions.
Collapse
Affiliation(s)
- Marta Segarra
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany; , .,Institute of Cell Biology and Neuroscience, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Maria R Aburto
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany; , .,Institute of Cell Biology and Neuroscience, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Jasmin Hefendehl
- Neurovascular Disorders, Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany.,Institute of Cell Biology and Neuroscience, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany; , .,Institute of Cell Biology and Neuroscience, University of Frankfurt, D-60438 Frankfurt am Main, Germany.,Max Planck Institute for Brain Research, D-60438 Frankfurt am Main, Germany
| |
Collapse
|
20
|
Transcriptome-wide Sequencing Reveals Molecules and Pathways Involved in Neurofibromatosis Type I Combined With Spinal Deformities. Spine (Phila Pa 1976) 2020; 45:E489-E498. [PMID: 31770328 DOI: 10.1097/brs.0000000000003338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
UNLABELLED MINI: We identified differentially expressed genes (DEGs) that may be involved in the development of neurofibromatosis type I by whole-transcriptional sequencing. Seven hundred eighty DEGs were identified which include protein coding genes, miRNAs, and lncRNAs. The enrichment analysis may reveal pathways that these DEGs involved. A total of 383 protein-pairs for DEGs may unfold the possible mechanism how the disease is developed. STUDY DESIGN This is a clinical basic study on neurofibromatosis type I (NF-1) with spinal deformity. OBJECTIVE The current research focuses on screening key molecules affecting NF-1 with spinal deformity by transcriptome sequencing and discovering its underlying molecular biological mechanisms. SUMMARY OF BACKGROUND DATA NF-1 is a complex multisystem human disorder, which is often found in spinal deformities patients. The success rate of orthopedic surgery for neurofibromatosis type I combined with spinal deformities patients was low because of the lack of molecular pathology. METHODS In our study, the transcriptome-wide sequencing was preformed to identify the differentially expressed genes (DEGs) involved in this disease. RESULTS Seven hundred eighty DEGs were identified which include protein coding genes, miRNAs, and lncRNAs. The DO, GO, KEGG and Reactome enrichment analysis may reveal pathways that these DEGs involved. And the 383 protein-pairs for DEGs that are involved in NF-1 combined with spinal deformities may unfold the possible mechanism how this disease is developed. CONCLUSION The differentially expressed miRNAs and lncRNAs may contribute the ceRNA network. We focused on three key DEGs: FGFR2, MAP3K1 and STAT4. FGFR2 and MAP3K1 are members of the RAS/RAF/MEK/ERK-signaling pathway, and STAT4 were involved in the JAK/STAT pathway. The expression changes were verified by other researches and the functional cross-talk between the Ras/MAPK and JAK/STAT pathways may contribute in the disease development. This study took insight of the molecular mechanism of this disease. More detailed interactions between these factors are needed to be further explored. These key DEGs and involved pathways may provide clues in the clinical process for patients with NF-1, especially in prognosis prediction. LEVEL OF EVIDENCE N/A.
Collapse
|
21
|
FGF2 and dual agonist of NCAM and FGF receptor 1, Enreptin, rescue neurite outgrowth loss in hippocampal neurons expressing mutated huntingtin proteins. J Neural Transm (Vienna) 2019; 126:1493-1500. [PMID: 31501979 DOI: 10.1007/s00702-019-02073-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/30/2019] [Indexed: 10/26/2022]
Abstract
In the present study, we developed an in vitro model of Huntington disease (HD) by transfecting primary rat hippocampal neurons with plasmids coding for m-htt exon 1 with different number of CAG repeats (18, 50 and 115) and demonstrated the influence of the length of polyQ sequence on neurite elongation. We found that exogenously applied FGF2 significantly rescued the m-htt-induced loss of neurite outgrowth. Moreover, the Enreptin peptide, an FGFR1 and NCAM dual agonist, had a similar neuritogenic effect to FGF2 in clinically relevant m-htt 50Q-expressing neurons. This study has developed an in vitro model of primary hippocampal neurons transfected with m-htt-coding vectors that is a powerful tool to study m-htt-related effects on neuronal placticity.
Collapse
|
22
|
Yuan H, Ni X, Zheng M, Han X, Song Y, Yu M. Effect of catalpol on behavior and neurodevelopment in an ADHD rat model. Biomed Pharmacother 2019; 118:109033. [PMID: 31545235 DOI: 10.1016/j.biopha.2019.109033] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 10/26/2022] Open
Abstract
Studies suggest that abnormal neurodevelopment of prefrontal striatal circuits is implicated in the pathogenesis of attention deficit hyperactivity disorder (ADHD). In the present study, we investigated the effect of catalpol, an active ingredient of Rehmanniae radix preparata, which is the most frequently used Chinese medicinal herb for the treatment of ADHD, on behavior and neurodevelopment in spontaneously hypertensive rats (SHR). SHR were divided into SHR group (vehicle, i.g.), methylphenidate (MPH) group (2 mg/kg/day, i.g.), and catalpol group (50 mg/kg/day i.g.), and Wistar-Kyoto (WKY) rats were used as control group (vehicle, i.g.). Open Field Test (OFT) and Morris water maze (MWM) test were performed to assess the effect of catalpol on behavior. Results revealed that both catalpol and MPH treatment decreased average speed, time spent in the central area, rearing times, and central area visits, increased the immobility time of SHR in OFT, and increased number of visits to the annulus, and time spent in target quadrant in the MWM test. Hematoxylin and eosin (H&E) staining showed that catalpol reduced irregular neuronal arrangement, ruptured nuclear membranes, and resulted in disappearance of the nucleolus in the prefrontal cortex (PFC) and striatum of SHR. Moreover, immuno-fluorescent staining of NeuN and myelin basic protein (MBP) indicated that catalpol ameliorated neuronal loss and contributed to myelination. Finally, western blot and immunostaining analysis suggested that several regulatory proteins involved in PFC development were up-regulated by catalpol treatment, such as brain-derived neurotrophic factor (BDNF), cyclin-dependent kinase 5 (Cdk5), p35, fibroblast growth factor (FGF) 21 and its receptor (FGFR)1. Taken together, catalpol can effectively ameliorate hyperactive and impulsive behavior, improve spatial learning and memory in SHR, likely through the neurodevelopmental pathways. Nonetheless, whether catalpol could attenuate inattention in SHR and the pathway by which catalpol reduces neuronal loss remain to be further studied.
Collapse
Affiliation(s)
- Haixia Yuan
- Institute of Pediatrics of traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Xinqiang Ni
- Pediatrics of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518038, Guangdong Province, China.
| | - Min Zheng
- Pediatrics of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518038, Guangdong Province, China.
| | - Xinmin Han
- Institute of Pediatrics of traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Yuchen Song
- Institute of Pediatrics of traditional Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Minfeng Yu
- Pediatrics of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518038, Guangdong Province, China.
| |
Collapse
|
23
|
Sassone J, Papadimitriou E, Thomaidou D. Regenerative Approaches in Huntington's Disease: From Mechanistic Insights to Therapeutic Protocols. Front Neurosci 2018; 12:800. [PMID: 30450032 PMCID: PMC6224350 DOI: 10.3389/fnins.2018.00800] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/15/2018] [Indexed: 01/10/2023] Open
Abstract
Huntington’s Disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the exon-1 of the IT15 gene encoding the protein Huntingtin. Expression of mutated Huntingtin in humans leads to dysfunction and ultimately degeneration of selected neuronal populations of the striatum and cerebral cortex. Current available HD therapy relies on drugs to treat chorea and control psychiatric symptoms, however, no therapy has been proven to slow down disease progression or prevent disease onset. Thus, although 24 years have passed since HD gene identification, HD remains a relentless progressive disease characterized by cognitive dysfunction and motor disability that leads to death of the majority of patients, on average 10–20 years after its onset. Up to now several molecular pathways have been implicated in the process of neurodegeneration involved in HD and have provided potential therapeutic targets. Based on these data, approaches currently under investigation for HD therapy aim on the one hand at getting insight into the mechanisms of disease progression in a human-based context and on the other hand at silencing mHTT expression by using antisense oligonucleotides. An innovative and still poorly investigated approach is to identify new factors that increase neurogenesis and/or induce reprogramming of endogenous neuroblasts and parenchymal astrocytes to generate new healthy neurons to replace lost ones and/or enforce neuroprotection of pre-existent striatal and cortical neurons. Here, we review studies that use human disease-in-a-dish models to recapitulate HD pathogenesis or are focused on promoting in vivo neurogenesis of endogenous striatal neuroblasts and direct neuronal reprogramming of parenchymal astrocytes, which combined with neuroprotective protocols bear the potential to re-establish brain homeostasis lost in HD.
Collapse
Affiliation(s)
- Jenny Sassone
- Vita-Salute University and San Raffaele Scientific Institute, Milan, Italy
| | | | - Dimitra Thomaidou
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
24
|
Wang L, Li XX, Chen X, Qin XY, Kardami E, Cheng Y. Antidepressant-Like Effects of Low- and High-Molecular Weight FGF-2 on Chronic Unpredictable Mild Stress Mice. Front Mol Neurosci 2018; 11:377. [PMID: 30369869 PMCID: PMC6194172 DOI: 10.3389/fnmol.2018.00377] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/21/2018] [Indexed: 12/11/2022] Open
Abstract
The occurrence of depressive disorder has long been attributed to changes in monoamines, with the focus of drug treatment strategies being to change the effectiveness of monoamines. However, the success achieved by changing these processes is limited and further stimulates the exploration of alternative mechanisms and treatments. Fibroblast growth factor 2 (FGF-2), which occurs in a high-molecular weight (HMW) and low-molecular weight (LMW) form, is a potent developmental modulator and nervous system regulator that has been suggested to play an important role in various psychiatric disorders. In this study, we investigated the antidepressant effects of HMW and LMW FGF-2 on depression induced by chronic stress. Both peripheral LMW and HMW FGF-2 attenuated the depression-like behaviors in chronic unpredictable mild stress (CUMS) mice to a similar extent, as determined by the forced swimming, tail suspension, and sucrose preference tests. We then showed that CUMS-induced oxidative stresses in mice were inhibited by FGF-2 treatments both in central and peripheral. We also showed that both forms of FGF-2 increased the phosphorylation of ERK and AKT, increased Bcl-2 expression and inhibited caspase-3 activation in CUMS mice. Interestingly, HMW FGF-2 enhanced the activity of the brain-derived neurotrophic factor (BDNF) to a greater extent than did LMW FGF-2 in the hippocampus. Taken together, these results suggest that depressive symptoms can be relieved by administering different forms of FGF-2 peripherally in a CUMS-induced depression model through a similar antidepressant signaling pathway, therefore suggesting a potential clinical use for FGF-2 as a treatment for depression.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Xi-Xi Li
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Xi Chen
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Xiao-Yan Qin
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Elissavet Kardami
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Yong Cheng
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| |
Collapse
|
25
|
|
26
|
Yamakawa M, Doh SJ, Santosa SM, Montana M, Qin EC, Kong H, Han KY, Yu C, Rosenblatt MI, Kazlauskas A, Chang JH, Azar DT. Potential lymphangiogenesis therapies: Learning from current antiangiogenesis therapies-A review. Med Res Rev 2018. [PMID: 29528507 DOI: 10.1002/med.21496] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In recent years, lymphangiogenesis, the process of lymphatic vessel formation from existing lymph vessels, has been demonstrated to have a significant role in diverse pathologies, including cancer metastasis, organ graft rejection, and lymphedema. Our understanding of the mechanisms of lymphangiogenesis has advanced on the heels of studies demonstrating vascular endothelial growth factor C as a central pro-lymphangiogenic regulator and others identifying multiple lymphatic endothelial biomarkers. Despite these breakthroughs and a growing appreciation of the signaling events that govern the lymphangiogenic process, there are no FDA-approved drugs that target lymphangiogenesis. In this review, we reflect on the lessons available from the development of antiangiogenic therapies (26 FDA-approved drugs to date), review current lymphangiogenesis research including nanotechnology in therapeutic drug delivery and imaging, and discuss molecules in the lymphangiogenic pathway that are promising therapeutic targets.
Collapse
Affiliation(s)
- Michael Yamakawa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Susan J Doh
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Samuel M Santosa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Mario Montana
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Ellen C Qin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Charles Yu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL.,Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
27
|
Lixing X, zhouye J, Liting G, Ruyi Z, Rong Q, Shiping M. Saikosaponin- d -mediated downregulation of neurogenesis results in cognitive dysfunction by inhibiting Akt/Foxg-1 pathway in mice. Toxicol Lett 2018; 284:79-85. [DOI: 10.1016/j.toxlet.2017.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/30/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022]
|
28
|
Gurung HR, Carr MM, Bryant K, Chucair-Elliott AJ, Carr DJJ. Fibroblast growth factor-2 drives and maintains progressive corneal neovascularization following HSV-1 infection. Mucosal Immunol 2018; 11:172-185. [PMID: 28378806 PMCID: PMC5628112 DOI: 10.1038/mi.2017.26] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/24/2017] [Indexed: 02/04/2023]
Abstract
Herpes simplex virus type 1 (HSV-1) infection of the cornea induces vascular endothelial growth factor A (VEGF-A)-dependent lymphangiogenesis that continues to develop well beyond the resolution of infection. Inflammatory leukocytes infiltrate the cornea and have been implicated to be essential for corneal neovascularization, an important clinically relevant manifestation of stromal keratitis. Here we report that cornea infiltrating leukocytes including neutrophils and T cells do not have a significant role in corneal neovascularization past virus clearance. Antibody-mediated depletion of these cells did not impact lymphatic or blood vessel genesis. Multiple pro-angiogenic factors including IL-6, angiopoietin-2, hepatocyte growth factor, fibroblast growth factor-2 (FGF-2), VEGF-A, and matrix metalloproteinase-9 were expressed within the cornea following virus clearance. A single bolus of dexamethasone at day 10 post infection (pi) resulted in suppression of blood vessel genesis and regression of lymphatic vessels at day 21 pi compared to control-treated mice. Whereas IL-6 neutralization had a modest impact on hemangiogenesis (days 14-21 pi) and lymphangiogenesis (day 21 pi) in a time-dependent fashion, neutralization of FGF-2 had a more pronounced effect on the suppression of neovascularization (blood and lymphatic vessels) in a time-dependent, leukocyte-independent manner. Furthermore, FGF-2 neutralization suppressed the expression of all pro-angiogenic factors measured and preserved visual acuity.
Collapse
Affiliation(s)
- Hem R. Gurung
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States of America
| | - Meghan M. Carr
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, United States of America
| | - Katie Bryant
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, United States of America
| | - Ana J. Chucair-Elliott
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, United States of America
| | - Daniel J. J. Carr
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States of America,Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, United States of America,Corresponding author:, Department of Ophthalmology, DMEI #A415, OUHSC, 608 Stanton L.Young Blvd., Oklahoma City, OK. 73104 USA, Phone: 405-271-8784
| |
Collapse
|
29
|
Wang Z. Experimental and Clinical Strategies for Treating Spinocerebellar Ataxia Type 3. Neuroscience 2017; 371:138-154. [PMID: 29229556 DOI: 10.1016/j.neuroscience.2017.11.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/02/2023]
Abstract
Spinocerebellar ataxia type 3 (SCA3), or Machado-Joseph disease (MJD), is an autosomal dominant neurodegenerative disorder caused by the expansion of a polyglutamine (polyQ) tract in the ataxin-3 protein. To date, there is no effective therapy available to prevent progression of this disease. However, clinical strategies for alleviating various symptoms are imperative to promote a better quality of life for SCA3/MJD patients. Furthermore, experimental therapeutic strategies, including gene silencing or mutant protein clearance, mutant polyQ protein modification, stabilizing the native protein conformation, rescue of cellular dysfunction and neuromodulation to slow the progression of SCA3/MJD, have been developed. In this study, based on the current knowledge, I detail the clinical and experimental therapeutic strategies for treating SCA3/MJD, paying particular attention to drug discovery.
Collapse
Affiliation(s)
- Zijian Wang
- Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an, Shaanxi 710065, China.
| |
Collapse
|
30
|
Connor B. Concise Review: The Use of Stem Cells for Understanding and Treating Huntington's Disease. Stem Cells 2017; 36:146-160. [PMID: 29178352 DOI: 10.1002/stem.2747] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/13/2017] [Indexed: 12/20/2022]
Abstract
Two decades ago, researchers identified that a CAG expansion mutation in the huntingtin (HTT) gene was involved in the pathogenesis of Huntington's disease (HD). However, since the identification of the HTT gene, there has been no advance in the development of therapeutic strategies to prevent or reduce the progression of HD. With the recent advances in stem cell biology and human cell reprogramming technologies, several novel and exciting pathways have emerged allowing researchers to enhance their understanding of the pathogenesis of HD, to identify and screen potential drug targets, and to explore alternative donor cell sources for cell replacement therapy. This review will discuss the role of compensatory neurogenesis in the HD brain, the use of stem cell-based therapies for HD to replace or prevent cell loss, and the recent advance of cell reprogramming to model and/or treat HD. These new technologies, coupled with advances in genome editing herald a promising new era for HD research with the potential to identify a therapeutic strategy to alleviate this debilitating disorder. Stem Cells 2018;36:146-160.
Collapse
Affiliation(s)
- Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Dzidziguri D, Modebadze I, Bakuradze E, Mosidze G, Berulava M. Determination of The Properties of Rat Brain Thermostable Protein Complex which Inhibit Cell Proliferation. CELL JOURNAL 2017; 19:552-558. [PMID: 29105389 PMCID: PMC5672093 DOI: 10.22074/cellj.2018.4835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/24/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Cell proliferation is known to be controlled by many networks of regulatory proteins. These multiple and complicated mechanisms of control are still being investigated. The aim of the present study is to determine the different properties of adult rat brain thermostable protein complex (TPC) which affect cell proliferation. MATERIALS AND METHODS This experimental study used brain, kidney and liver tissue from adult (150-170 g) and adolescent (7, 10, 21, 28 days) white rats, adult pigeons and mice. Brain TPC was isolated by alcohol extraction, and primary antibodies Ki67 and GAD65/67 were used for immunohistochemistry, evaluation of transcriptional activity of the tissues and determination of the mitotic index. RESULTS The results show that brain TPC from rats reversibly decreases cell proliferation by inhibiting transcription. The evidence suggests that TPC is not species-specific, but expresses tissue specificity with regards to terminally differentiated cells. Rat brain TPC inhibits mitotic activity of the progenitor cells in the dentate gyrus of adolescent rats, and corresponding with this decrease in the mitotic index the number of Ki67 positive cells increases. Simultaneously, the number of GAD65/67-positive cells in the hippocampus decreases by approximately threefold. CONCLUSIONS These results indicate that rat brain TPC causes the reversible suppression of cell proliferation through the inhibition of transcription. Inhibitory effects of rat brain TPC leads to an increase the number of cells in the cell cycle, in tissues of adolescent rats.
Collapse
Affiliation(s)
- Diana Dzidziguri
- Department of Biology, Faculty of Exact and Natural Sciences, Iv. Javakhishvili Tbilisi State University, Tbilisi, Georgia. Electronic address :
| | - Irina Modebadze
- Department of Biology, Faculty of Exact and Natural Sciences, Iv. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Ekaterine Bakuradze
- Department of Biology, Faculty of Exact and Natural Sciences, Iv. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Giorgi Mosidze
- Department of Biology, Faculty of Exact and Natural Sciences, Iv. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Manana Berulava
- Department of Biology, Faculty of Exact and Natural Sciences, Iv. Javakhishvili Tbilisi State University, Tbilisi, Georgia.,Faculty of Natural Sciences and Healthcare, Sokhumi State University, Tbilisi, Georgia
| |
Collapse
|
32
|
Tang MM, Lin WJ, Zhang JT, Zhao YW, Li YC. Exogenous FGF2 reverses depressive-like behaviors and restores the suppressed FGF2-ERK1/2 signaling and the impaired hippocampal neurogenesis induced by neuroinflammation. Brain Behav Immun 2017; 66:322-331. [PMID: 28529071 DOI: 10.1016/j.bbi.2017.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/10/2017] [Accepted: 05/17/2017] [Indexed: 01/03/2023] Open
Abstract
Our previous work demonstrated that neuroinflammation evoked by triple repeated central LPS challenges inhibited adult hippocampal neurogenesis that were correlated with the depressive-like behavioral symptoms induced by neuroinflammation. These findings suggest that hippocampal neurogenesis might be one of biological mechanisms underlying depression induced by neuroinflammation and targeting neurogenesis might lead to new therapeutic strategies for the treatment of depression. In this study, we manipulated adult hippocampal neurogenesis using fibroblast growth factor 2 (FGF2), one crucial molecule modulating cell proliferation and survival in central nervous system, and investigate the involvement and the potential therapeutic effects of FGF2 on neuroinflammation-induced depression. Central lipopolysaccharides (LPS) challenges were used as previously to evoke the neuroinflammatory state in the brain of rat. Exogenous FGF2 was infused into lateral ventricle during the neuroinflammatory state. It was found that the protein expression of FGF2 in hippocampus was inhibited by neuroinflammation. The activation of extracellular signal-regulated kinase (ERK), the downstream molecule of FGF2, was also inhibited by neuroinflammation. Exogenous FGF2 infusions prevented the decrease in phosphorylation of ERK1/2 under neuroinflammation state. Exogenous FGF2 reversed depressive-like behaviors and the impaired hippocampal neurogenesis induced by neuroinflammation. These findings provide evidence that the FGF2-ERK1/2 pathway is involved in the pathophysiology of depressive-like behaviors, and manipulating the neurogenesis pathway is a viable therapeutic approach to inflammation-associated depression.
Collapse
Affiliation(s)
- Ming-Ming Tang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wen-Juan Lin
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China; Brain-Behavior Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jun-Tao Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Wei Zhao
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying-Cong Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
| |
Collapse
|
33
|
Grade S, Götz M. Neuronal replacement therapy: previous achievements and challenges ahead. NPJ Regen Med 2017; 2:29. [PMID: 29302363 PMCID: PMC5677983 DOI: 10.1038/s41536-017-0033-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022] Open
Abstract
Lifelong neurogenesis and incorporation of newborn neurons into mature neuronal circuits operates in specialized niches of the mammalian brain and serves as role model for neuronal replacement strategies. However, to which extent can the remaining brain parenchyma, which never incorporates new neurons during the adulthood, be as plastic and readily accommodate neurons in networks that suffered neuronal loss due to injury or neurological disease? Which microenvironment is permissive for neuronal replacement and synaptic integration and which cells perform best? Can lost function be restored and how adequate is the participation in the pre-existing circuitry? Could aberrant connections cause malfunction especially in networks dominated by excitatory neurons, such as the cerebral cortex? These questions show how important connectivity and circuitry aspects are for regenerative medicine, which is the focus of this review. We will discuss the impressive advances in neuronal replacement strategies and success from exogenous as well as endogenous cell sources. Both have seen key novel technologies, like the groundbreaking discovery of induced pluripotent stem cells and direct neuronal reprogramming, offering alternatives to the transplantation of fetal neurons, and both herald great expectations. For these to become reality, neuronal circuitry analysis is key now. As our understanding of neuronal circuits increases, neuronal replacement therapy should fulfill those prerequisites in network structure and function, in brain-wide input and output. Now is the time to incorporate neural circuitry research into regenerative medicine if we ever want to truly repair brain injury.
Collapse
Affiliation(s)
- Sofia Grade
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
34
|
Yang L, Hansen Falkesgaard M, Thulstrup PW, Walmod PS, Lo Leggio L, Krighaar Rasmussen K. Expression, refolding and spectroscopic characterization of fibronectin type III (FnIII)-homology domains derived from human fibronectin leucine rich transmembrane protein (FLRT)-1, -2, and -3. PeerJ 2017; 5:e3550. [PMID: 28698826 PMCID: PMC5502089 DOI: 10.7717/peerj.3550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/15/2017] [Indexed: 12/28/2022] Open
Abstract
The fibronectin leucine rich transmembrane (FLRT) protein family consists in humans of 3 proteins, FLRT1, -2, and -3. The FLRT proteins contain two extracellular domains separated by an unstructured linker. The most membrane distal part is a leucine rich repeat (LRR) domain responsible for both cis- and trans-interactions, whereas the membrane proximal part is a fibronectin type III (FnIII) domain responsible for a cis-interaction with members of the fibroblast growth factor receptor 1 (FGFR1) family, which results in FGFR tyrosine kinase activation. Whereas the structures of FLRT LRR domains from various species have been determined, the expression and purification of recombinant FLRT FnIII domains, important steps for further structural and functional characterizations of the proteins, have not yet been described. Here we present a protocol for expressing recombinant FLRT-FnIII domains in inclusion bodies in Escherichia coli. His-tags permitted affinity purification of the domains, which subsequently were refolded on a Ni-NTA agarose column by reducing the concentration of urea. The refolding was confirmed by circular dichroism (CD) and 1H-NMR. By thermal unfolding experiments we show that a strand-strand cystine bridge has significant effect on the stability of the FLRT FnIII fold. We further show by Surface Plasmon Resonance that all three FnIII domains bind to FGFR1, and roughly estimate a Kd for each domain, all Kds being in the µM range.
Collapse
Affiliation(s)
- Lila Yang
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Maria Hansen Falkesgaard
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Waaben Thulstrup
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Peter Schledermann Walmod
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leila Lo Leggio
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Kim Krighaar Rasmussen
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark.,Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Gao C, Wang Q, Chung SK, Shen J. Crosstalk of metabolic factors and neurogenic signaling in adult neurogenesis: Implication of metabolic regulation for mental and neurological diseases. Neurochem Int 2017; 106:24-36. [DOI: 10.1016/j.neuint.2017.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/10/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022]
|
36
|
Acupuncture Stimulation at GB34 Restores MPTP-Induced Neurogenesis Impairment in the Subventricular Zone of Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:3971675. [PMID: 28592981 PMCID: PMC5448055 DOI: 10.1155/2017/3971675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/07/2017] [Accepted: 04/26/2017] [Indexed: 01/12/2023]
Abstract
Adult neurogenesis has recently been considered a new therapeutic paradigm of Parkinson's disease. In this study, we investigated whether acupuncture restores 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- (MPTP-) induced impaired neurogenesis in the subventricular zone (SVZ). Male C57BL/6 mice were given 30 mg/kg of MPTP intraperitoneally once a day for 5 days, after which they were intraperitoneally injected with 50 mg/kg of bromodeoxyuridine (BrdU) and given acupuncture stimulation at HT7 or GB34 for 12 consecutive days. Dopaminergic neuronal survival in the nigrostriatal pathway and cell proliferation in the SVZ was then evaluated by immunostaining. MPTP administration induced dopaminergic neuronal death in the nigrostriatal pathway, which was suppressed by acupuncture stimulation at GB34. MPTP administration also suppressed the number of BrdU-positive cells and glial fibrillary acidic protein/BrdU-positive cells and increased the number of doublecortin/BrdU-positive cells in the SVZ, which were restored by acupuncture stimulation at GB34. These results indicate that acupuncture stimulation at GB34 restores MPTP-induced neurogenesis impairment.
Collapse
|
37
|
Kotapalli SS, Dasari C, Duscharla D, Kami Reddy KR, Kasula M, Ummanni R. All-Trans-Retinoic Acid Stimulates Overexpression of Tumor Protein D52 (TPD52, Isoform 3) and Neuronal Differentiation of IMR-32 Cells. J Cell Biochem 2017; 118:4358-4369. [PMID: 28436114 DOI: 10.1002/jcb.26090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/21/2017] [Indexed: 11/06/2022]
Abstract
Tumor protein D52 (TPD52), a proto-oncogene is overexpressed in a variety of epithelial carcinomas and plays an important role in cell proliferation, migration, and cell death. In the present study we found that the treatment of IMR-32 neuroblastoma (NB) cells with retinoic acid (RA) stimulates an increase in expression of TPD52. TPD52 expression is detectable after 72 h, can be maintained till differentiation of NB cells suggesting that TPD52 is involved in differentiation. Here, we demonstrate that TPD52 is essential for RA to promote differentiation of NB cells. Our results show that exogenous expression of EGFP-TPD52 in IMR-32 cells resulted cell differentiation even without RA. RA by itself and with overexpression of TPD52 can increase the ability of NB cells differentiation. Interestingly, transfection of IMR-32 cells with a specific small hairpin RNA for efficient knockdown of TPD52 attenuated RA induced NB cells differentiation. Transcriptional and translational level expression of neurotropic (BDNF, NGF, Nestin) and differentiation (β III tubulin, NSE, TH) factors in NB cells with altered TPD52 expression and/or RA treatment confirmed essential function of TPD52 in cellular differentiation. Furthermore, we show that TPD52 protects cells from apoptosis and arrest cell proliferation by varying expression of p27Kip1, activation of Akt and ERK1/2 thus promoting cell differentiation. Additionally, inhibition of STAT3 activation by its specific inhibitor arrested NB cells differentiation by EGFP-TPD52 overexpression with or without RA. Taken together, our data reveal that TPD52 act through activation of JAK/STAT signaling pathway to undertake NB cells differentiation induced by RA. J. Cell. Biochem. 118: 4358-4369, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sudha Sravanti Kotapalli
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Chandrashekhar Dasari
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.,Centre for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Divya Duscharla
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.,Centre for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Karthik Reddy Kami Reddy
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.,Centre for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Manjula Kasula
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Ramesh Ummanni
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India.,Centre for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| |
Collapse
|
38
|
Metabolic Factors and Adult Neurogenesis: Impacts of Chinese Herbal Medicine on Brain Repair in Neurological Diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 135:117-147. [PMID: 28807156 DOI: 10.1016/bs.irn.2017.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adult neurogenesis plays the important roles in animal cognitive and emotional behaviors. Abnormal proliferation and differentiation of neural stem cells (NSCs) usually associate with the neural dysfunctions induced by different brain disorders. Therefore, targeting neurogenic factors could be a promoting strategy for neural regeneration and brain repair. Importantly, epidemiological studies suggest metabolism disorders like diabetes and obesity significantly increase the risk of neurological and psychiatric diseases. A large number of studies indicate that metabolic factors could serve as the modulators to adult neurogenesis, providing the potentials of metabolic factors to regulate NSCs growth and neural regeneration therapy. This chapter reviews the current studies on the roles of metabolic factors in modulating adult neurogenesis and evaluates the potentials of Chinese Herbal Medicine (CHM) for the treatment of neurological or psychiatric disorders by targeting the metabolic factors. Traditional Chinese Medicine (TCM) including CHM and acupuncture is now widely applied for the treatment of metabolic diseases, and neurological diseases in Asia, because its' therapeutic principles meet the multiple targets and complexity characteristics of most neurological disorders. Different studies indicate that there are many active compounds perform the regulations to metabolic factors and promoting neurogenesis. This chapter systematically summarizes the current progress and understanding of the active compounds and their underlying mechanisms of CHM formulas for promoting neurogenesis. Many CHM formulas and their active ingredients that originally used for metabolic disorders show the promising effects on mediating neurogenesis and brain repair for the treatments of neurodegenerative diseases. Therefore, further investigations about the relationship between neurogenesis and metabolic regulations of CHM will bring new insights into understanding the mechanisms of adult neurogenesis and provide great opportunities to develop new therapeutic strategies for neurological diseases. Those studies will provide scientific guidance to develop the drugs from TCM resource.
Collapse
|
39
|
Abdanipour A, Noori-Zadeh A, Mesbah-Namin SA, Bakhtiyari S, Nejatbakhsh R, Anarkooli IJ. Di-(2-ethylhexyl) Phthalate-Induced Hippocampus-Derived Neural Stem Cells Proliferation. CELL JOURNAL 2016; 19:166-172. [PMID: 28367427 PMCID: PMC5241513 DOI: 10.22074/cellj.2016.4862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
The brain and spinal cord have a limited capacity for self-repair under damaged conditions. One of the best options to overcome these limitations involves the use of phytochemicals as potential therapeutic agents. In this study, we have aimed to investigate the
effects of di-(2-ethylhexyl) phthalate (DEHP) on hippocampus-derived neural stem cells
(NSCs) proliferation to search phytochemical candidates for possible treatment of neurological diseases using endogenous capacity.
In this experimental study, neonatal rat hippocampus-derived NSCs were cultured and
treated with various concentrations of DEHP (0, 100, 200, 400 and 600 µM) and Cirsium
vulgare (C. vulgare) hydroethanolic extract (0, 200, 400, 600, 800 and 1000 µg/ml) for 48
hours under in vitro conditions. Cell proliferation rates and quantitative Sox2 gene expression were evaluated using MTT assay and real-time reverse transcription polymerase
chain reaction (RT-PCR).
We observed the highest average growth rate in the 400 µM DEHP and 800 µg/ml C.
vulgare extract treated groups. Sox2 expression in the DEHP-treated NSCs significantly
increased compared to the control group. Gas chromatography/mass spectrometry (GC/
MS) results demonstrated that the active ingredients that naturally occurred in the C. vulgare hydroethanolic extract were 2-ethyl-1-hexanamine, n-heptacosane, 1-cyclopentanecarboxylic acid, 1-heptadecanamine, 2,6-octadien-1-ol,2,6,10,14,18,22-tetracosahexaene, and DEHP. DEHP profoundly stimulated NSCs proliferation through Sox2 gene
overexpression.
These results provide and opportunity for further use of the C. vulgure phytochemicals for
prevention and/or treatment of neurological diseases via phytochemical mediated-proliferation of endogenous adult NSCs.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Noori-Zadeh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Salar Bakhtiyari
- Student Research Committee Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Reza Nejatbakhsh
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Iraj Jafari Anarkooli
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
40
|
Esteves S, Duarte-Silva S, Maciel P. Discovery of Therapeutic Approaches for Polyglutamine Diseases: A Summary of Recent Efforts. Med Res Rev 2016; 37:860-906. [PMID: 27870126 DOI: 10.1002/med.21425] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/01/2016] [Accepted: 10/05/2016] [Indexed: 12/19/2022]
Abstract
Polyglutamine (PolyQ) diseases are a group of neurodegenerative disorders caused by the expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the coding region of specific genes. This leads to the production of pathogenic proteins containing critically expanded tracts of glutamines. Although polyQ diseases are individually rare, the fact that these nine diseases are irreversibly progressive over 10 to 30 years, severely impairing and ultimately fatal, usually implicating the full-time patient support by a caregiver for long time periods, makes their economic and social impact quite significant. This has led several researchers worldwide to investigate the pathogenic mechanism(s) and therapeutic strategies for polyQ diseases. Although research in the field has grown notably in the last decades, we are still far from having an effective treatment to offer patients, and the decision of which compounds should be translated to the clinics may be very challenging. In this review, we provide a comprehensive and critical overview of the most recent drug discovery efforts in the field of polyQ diseases, including the most relevant findings emerging from two different types of approaches-hypothesis-based candidate molecule testing and hypothesis-free unbiased drug screenings. We hereby summarize and reflect on the preclinical studies as well as all the clinical trials performed to date, aiming to provide a useful framework for increasingly successful future drug discovery and development efforts.
Collapse
Affiliation(s)
- Sofia Esteves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| |
Collapse
|
41
|
Zhou S, Ochalek A, Szczesna K, Avci HX, Kobolák J, Varga E, Rasmussen M, Holst B, Cirera S, Hyttel P, Freude KK, Dinnyés A. The positional identity of iPSC-derived neural progenitor cells along the anterior-posterior axis is controlled in a dosage-dependent manner by bFGF and EGF. Differentiation 2016; 92:183-194. [PMID: 27321088 DOI: 10.1016/j.diff.2016.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 05/30/2016] [Accepted: 06/09/2016] [Indexed: 12/27/2022]
Abstract
Neural rosettes derived from human induced pluripotent stem cells (iPSCs) have been claimed to be a highly robust in vitro cellular model for biomedical application. They are able to propagate in vitro in the presence of mitogens, including basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF). However, these two mitogens are also involved in anterior-posterior patterning in a gradient dependent manner along the neural tube axis. Here, we compared the regional identity of neural rosette cells and specific neural subtypes of their progeny propagated with low and high concentrations of bFGF and EGF. We observed that low concentrations of bFGF and EGF in the culturing system were able to induce forebrain identity of the neural rosettes and promote subsequent cortical neuronal differentiation. On the contrary, high concentrations of these mitogens stimulate a mid-hindbrain fate of the neural rosettes, resulting in subsequent cholinergic neuron differentiation. Thus, our results indicate that different concentrations of bFGF and EGF supplemented during propagation of neural rosettes are involved in altering the identity of the resultant neural cells.
Collapse
Affiliation(s)
- Shuling Zhou
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anna Ochalek
- BioTalentum Ltd., Gödöllő, Hungary; Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllő, Hungary.
| | | | - Hasan X Avci
- BioTalentum Ltd., Gödöllő, Hungary; Department of Medical Chemistry, University of Szeged, Dóm tér 8, 6720 Szeged, Hungary.
| | | | - Eszter Varga
- BioTalentum Ltd., Gödöllő, Hungary; Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllő, Hungary.
| | | | | | - Susanna Cirera
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Kristine K Freude
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - András Dinnyés
- BioTalentum Ltd., Gödöllő, Hungary; Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllő, Hungary; Departments of Equine Sciences and Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
42
|
Dunbar GL, Sandstrom MI, Rossignol J, Lescaudron L. Neurotrophic Enhancers as Therapy for Behavioral Deficits in Rodent Models of Huntington's Disease: Use of Gangliosides, Substituted Pyrimidines, and Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2016; 5:63-79. [PMID: 16801683 DOI: 10.1177/1534582306289367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The interest in using neurotrophic factors as potential treatments for neurodegenerative disorders, such as Huntington's disease, has grown in the past decade. A major impediment for the clinical utility of neurotrophic factors is their inability to cross the blood-brain barrier in therapeutically significant amounts. Although several novel mechanisms for delivering exogenous neurotrophins to the brain have been developed, most of them involve invasive procedures or present significant risks. One approach to circumventing these problems is using therapeutic agents that can be administered systemically and have the ability to enhance the activity of neurotrophic factors. This review highlights the use of gangliosides, substituted pyrimidines, and mesenchymal stem cells as neurotrophic enhancers that have significant therapeutic potential while avoiding the pitfalls of delivering exogenous neurotrophic factors through the blood-brain barrier. The review focuses on the potential of these neurotrophic enhancers for treating the behavioral deficits in rodent models of Huntington's disease.
Collapse
|
43
|
Rodella LF, Rezzani R, Bonomini F, Peroni M, Cocchi MA, Hirtler L, Bonardelli S. Abdominal aortic aneurysm and histological, clinical, radiological correlation. Acta Histochem 2016; 118:256-62. [PMID: 26858185 DOI: 10.1016/j.acthis.2016.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 01/26/2016] [Accepted: 01/28/2016] [Indexed: 10/22/2022]
Abstract
To date, the pathogenesis of abdominal aortic aneurism (AAA) still remains unclear. As such, the aim of this study was to evaluate changes of the aortic structure during AAA. We analysed the microscopic frame of vessels sections, starting from the primum movens leading to abnormal dilatation. AAA samples were collected and processed through various staining methods (Verhoeff-Van Gieson, Masson Goldner, Sirius Red). Subsequently, the vessel morphology and collagenic web of the tunica media and adventitia were determined and the amount of type I and type III collagen was measured. We also applied immune-histochemistry markers for CD34 and PGP 9.5 in order to identify vascular and nerve structures in the aorta. Immune-positivity quantification was used to calculate the percentage of the stained area. We found increasing deposition of type I collagen and reduced type III collagen in both tunica media and adventitia of AAA. The total amount of vasa vasorum, marked with CD34, and nerva vasorum, marked with PGP 9.5, was also higher in AAA samples. Cardiovascular risk factors (blood pressure, dyslipidemia, cigarette smoking) and radiological data (maximum aneurism diameter, intra-luminal thrombus, aortic wall calcification) increased these changes. These results suggest that the tunica adventitia may have a central role in the pathogenesis of AAA as clearly there are major changes characterized by rooted inflammatory infiltration. The presence of immune components could explain these modifications within the framework of the aorta.
Collapse
|
44
|
Chandrasekaran S, Bonchev D. Network analysis of human post-mortem microarrays reveals novel genes, microRNAs, and mechanistic scenarios of potential importance in fighting huntington's disease. Comput Struct Biotechnol J 2016; 14:117-130. [PMID: 27924190 PMCID: PMC5128196 DOI: 10.1016/j.csbj.2016.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 01/28/2016] [Accepted: 02/02/2016] [Indexed: 01/18/2023] Open
Abstract
Huntington's disease is a progressive neurodegenerative disorder characterized by motor disturbances, cognitive decline, and neuropsychiatric symptoms. In this study, we utilized network-based analysis in an attempt to explore and understand the underlying molecular mechanism and to identify critical molecular players of this disease condition. Using human post-mortem microarrays from three brain regions (cerebellum, frontal cortex and caudate nucleus) we selected in a four-step procedure a seed set of highly modulated genes. Several protein-protein interaction networks, as well as microRNA-mRNA networks were constructed for these gene sets with the Elsevier Pathway Studio software and its associated ResNet database. We applied a gene prioritizing procedure based on vital network topological measures, such as high node connectivity and centrality. Adding to these criteria the guilt-by-association rule and exploring their innate biomolecular functions, we propose 19 novel genes from the analyzed microarrays, from which CEBPA, CDK1, CX3CL1, EGR1, E2F1, ERBB2, LRP1, HSP90AA1 and ZNF148 might be of particular interest for experimental validation. A possibility is discussed for dual-level gene regulation by both transcription factors and microRNAs in Huntington's disease mechanism. We propose several possible scenarios for experimental studies initiated via the extra-cellular ligands TGFB1, FGF2 and TNF aiming at restoring the cellular homeostasis in Huntington's disease.
Collapse
Affiliation(s)
- Sreedevi Chandrasekaran
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, VA, USA
| | | |
Collapse
|
45
|
Cai P, Ye J, Zhu J, Liu D, Chen D, Wei X, Johnson NR, Wang Z, Zhang H, Cao G, Xiao J, Ye J, Lin L. Inhibition of Endoplasmic Reticulum Stress is Involved in the Neuroprotective Effect of bFGF in the 6-OHDA-Induced Parkinson's Disease Model. Aging Dis 2016; 7:336-449. [PMID: 27493838 PMCID: PMC4963188 DOI: 10.14336/ad.2016.0117] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/17/2016] [Indexed: 01/20/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder with complicated pathophysiologic mechanisms. Endoplasmic reticulum (ER) stress appears to play a critical role in the progression of PD. We demonstrated that basic fibroblast growth factor (bFGF), as a neurotropic factor, inhibited ER stress-induced neuronal cell apoptosis and that 6-hydroxydopamine (6-OHDA)-induced ER stress was involved in the progression of PD in rats. bFGF administration improved motor function recovery, increased tyrosine hydroxylase (TH)-positive neuron survival, and upregulated the levels of neurotransmitters in PD rats. The 6-OHDA-induced ER stress response proteins were inhibited by bFGF treatment. Meanwhile, bFGF also increased expression of TH. The administration of bFGF activated the downstream signals PI3K/Akt and Erk1/2 in vivo and in vitro. Inhibition of the PI3K/Akt and Erk1/2 pathways by specific inhibitors partially reduced the protective effect of bFGF. This study provides new insight towards bFGF translational drug development for PD involving the regulation of ER stress.
Collapse
Affiliation(s)
- Pingtao Cai
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jingjing Ye
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jingjing Zhu
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Dan Liu
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Daqing Chen
- 2Emergency Department, the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaojie Wei
- 3Department of Neurosurgery, Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Noah R Johnson
- 4Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Zhouguang Wang
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hongyu Zhang
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Guodong Cao
- 4Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Jian Xiao
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Junming Ye
- 5Department of Anesthesia, the First Affiliated Hospital, Gannan Medical College, Ganzhou, 341000, China
| | - Li Lin
- 1School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| |
Collapse
|
46
|
Domouzoglou EM, Naka KK, Vlahos AP, Papafaklis MI, Michalis LK, Tsatsoulis A, Maratos-Flier E. Fibroblast growth factors in cardiovascular disease: The emerging role of FGF21. Am J Physiol Heart Circ Physiol 2015; 309:H1029-H1038. [PMID: 26232236 PMCID: PMC4747916 DOI: 10.1152/ajpheart.00527.2015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/22/2015] [Indexed: 01/07/2023]
Abstract
Early detection of risk factors for enhanced primary prevention and novel therapies for treating the chronic consequences of cardiovascular disease are of the utmost importance for reducing morbidity. Recently, fibroblast growth factors (FGFs) have been intensively studied as potential new molecules in the prevention and treatment of cardiovascular disease mainly attributable to metabolic effects and angiogenic actions. Members of the endocrine FGF family have been shown to increase metabolic rate, decrease adiposity, and restore glucose homeostasis, suggesting a multiple metabolic role. Serum levels of FGFs have been associated with established cardiovascular risk factors as well as with the severity and extent of coronary artery disease and could be useful for prediction of cardiovascular death. Furthermore, preclinical investigations and clinical trials have tested FGF administration for therapeutic angiogenesis in ischemic vascular disease, demonstrating a potential role in improving angina and limb function. FGF21 has lately emerged as a potent metabolic regulator with multiple effects that ultimately improve the lipoprotein profile. Early studies show that FGF21 is associated with the presence of atherosclerosis and may play a protective role against plaque formation by improving endothelial function. The present review highlights recent investigations suggesting that FGFs, in particular FGF21, may be useful as markers of cardiovascular risk and may also serve as protective/therapeutic agents in cardiovascular disease.
Collapse
Affiliation(s)
- Eleni M Domouzoglou
- Department of Pediatrics, Medical School, University of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- Second Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Antonios P Vlahos
- Department of Pediatrics, Medical School, University of Ioannina, Ioannina, Greece
| | - Michail I Papafaklis
- Second Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Lampros K Michalis
- Second Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Agathoklis Tsatsoulis
- Department of Endocrinology, Medical School, University of Ioannina, Ioannina, Greece
| | - Eleftheria Maratos-Flier
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Xie W, Wang JQ, Wang QC, Wang Y, Yao S, Tang TS. Adult neural progenitor cells from Huntington's disease mouse brain exhibit increased proliferation and migration due to enhanced calcium and ROS signals. Cell Prolif 2015; 48:517-31. [PMID: 26269226 DOI: 10.1111/cpr.12205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/10/2015] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Huntington's disease (HD) is an inherited human neurodegenerative disorder characterized by uncontrollable movement, psychiatric disturbance and cognitive decline. Impaired proliferative/differentiational potentials of adult neural progenitor cells (ANPCs) have been thought to be a pathogenic mechanism involved in it. In this study, we aimed to elucidate intrinsic properties of ANPCs subjected to neurodegenerative condition in YAC128 HD mice. MATERIALS AND METHODS ANPCs were isolated from the SVZ regions of 4-month-old WT and YAC128 mice. Cell proliferation, migration and neuronal differentiation in vitro were compared between these two genotypes with/without Ca(2+) inhibitors or ROS scavenger treatments. Differences in ANPC proliferation and differentiation capabilities in vivo between the two genotypes were evaluated using Ki-67 and Doublecortin (DCX) immunofluorescence respectively. RESULTS Compared to WT ANPCs, YAC128 ANPCs had significantly enhanced cell proliferation, migration and neuronal differentiation in vitro, accompanied by increased Ca(2+) and ROS signals. Raised proliferation and migration in YAC128 ANPCs were abolished by Ca(2+) signalling antagonists and ROS scavenging. However, in vivo, HD ANPCs failed to show any elevated proliferation or differentiation. CONCLUSIONS Increased Ca(2+) signalling and higher level of ROS conferred HD ANPC enhancement of proliferation and migration potentials. However, the in vivo micro-environment did not support endogenous ANPCs to respond appropriately to neuronal loss in these YAC128 mouse brains.
Collapse
Affiliation(s)
- Wenjuan Xie
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiu-Qiang Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiao-Chu Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yun Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Sheng Yao
- Department of Neurology, Navy General Hospital, Beijing, 100048, China
| | - Tie-Shan Tang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
48
|
Cheng Y, Rodriguiz RM, Murthy SRK, Senatorov V, Thouennon E, Cawley NX, Aryal DK, Ahn S, Lecka-Czernik B, Wetsel WC, Loh YP. Neurotrophic factor-α1 prevents stress-induced depression through enhancement of neurogenesis and is activated by rosiglitazone. Mol Psychiatry 2015; 20:744-54. [PMID: 25330741 PMCID: PMC4405386 DOI: 10.1038/mp.2014.136] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/14/2014] [Accepted: 09/04/2014] [Indexed: 12/24/2022]
Abstract
Major depressive disorder is often linked to stress. Although short-term stress is without effect in mice, prolonged stress leads to depressive-like behavior, indicating that an allostatic mechanism exists in this difference. Here we demonstrate that mice after short-term (1 h per day for 7 days) chronic restraint stress (CRS), do not display depressive-like behavior. Analysis of the hippocampus of these mice showed increased levels of neurotrophic factor-α1 (NF-α1; also known as carboxypeptidase E, CPE), concomitant with enhanced fibroblast growth factor 2 (FGF2) expression, and an increase in neurogenesis in the dentate gyrus. In contrast, after prolonged (6 h per day for 21 days) CRS, mice show decreased hippocampal NF-α1 and FGF2 levels and depressive-like responses. In NF-α1-knockout mice, hippocampal FGF2 levels and neurogenesis are reduced. These mice exhibit depressive-like behavior that is reversed by FGF2 administration. Indeed, studies in cultured hippocampal neurons reveal that NF-α1 treatment directly upregulates FGF2 expression through extracellular signal-regulated kinase-Sp1 signaling. Thus, during short-term CRS, hippocampal NF-α1 expression is upregulated and has a key role in preventing the onset of depressive-like behavior through enhanced FGF2-mediated neurogenesis. To evaluate the therapeutic potential of this pathway, we examined, rosiglitazone (Rosi), a PPARγ agonist, which has been shown to have antidepressant activity in rodents and humans. Rosi upregulates FGF2 expression in a NF-α1-dependent manner in hippocampal neurons. Mice fed Rosi show increased hippocampal NF-α1 levels and neurogenesis compared with controls, thereby indicating the antidepressant action of this drug. Development of drugs that activate the NF-α1/FGF2/neurogenesis pathway can offer a new approach to depression therapy.
Collapse
Affiliation(s)
- Yong Cheng
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramona M. Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Saravana R. K. Murthy
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vladimir Senatorov
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erwan Thouennon
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Niamh X. Cawley
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dipendra K. Aryal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Sohyun Ahn
- Program in Genomics Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beata Lecka-Czernik
- Departments of Orthopaedic Surgery and Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, Toledo, Ohio 43614, USA
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA,Departments of Neurobiology and Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Y. Peng Loh
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA,Correspondence should be addressed to: Dr. Y. Peng Loh, 49 Convent Drive, Bldg. 49, Room 6C80, National Institutes of Health, Bethesda, MD 20892, USA. , Tel: 301-496-3239, Fax: 301-496-9938
| |
Collapse
|
49
|
Embryonic neural stem cells in a 3D bioassay for trophic stimulation studies. Brain Res Bull 2015; 115:37-44. [DOI: 10.1016/j.brainresbull.2015.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 01/19/2023]
|
50
|
Chang R, Liu X, Li S, Li XJ. Transgenic animal models for study of the pathogenesis of Huntington's disease and therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2179-88. [PMID: 25931812 PMCID: PMC4404937 DOI: 10.2147/dddt.s58470] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Huntington’s disease (HD) is caused by a genetic mutation that results in polyglutamine expansion in the N-terminal regions of huntingtin. As a result, this polyQ expansion leads to the misfolding and aggregation of mutant huntingtin as well as age-dependent neurodegeneration. The genetic mutation in HD allows for generating a variety of animal models that express different forms of mutant huntingtin and show differential pathology. Studies of these animal models have provided an important insight into the pathogenesis of HD. Mouse models of HD include transgenic mice, which express N-terminal or full-length mutant huntingtin ubiquitously or selectively in different cell types, and knock-in mice that express full-length mutant Htt at the endogenous level. Large animals, such as pig, sheep, and monkeys, have also been used to generate animal HD models. This review focuses on the different features of commonly used transgenic HD mouse models as well as transgenic large animal models of HD, and also discusses how to use them to identify potential therapeutics. Since HD shares many pathological features with other neurodegenerative diseases, identification of therapies for HD would also help to develop effective treatment for different neurodegenerative diseases that are also caused by protein misfolding and occur in an age-dependent manner.
Collapse
Affiliation(s)
- Renbao Chang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xudong Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiao-Jiang Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People's Republic of China ; Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|