1
|
Sinha A, Nickerson G, Bouyain S, Matthews RT. Contactin-1 is a critical neuronal cell surface receptor for perineuronal net structure. J Biol Chem 2025; 301:108504. [PMID: 40220999 DOI: 10.1016/j.jbc.2025.108504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Perineuronal nets (PNNs) are neuron-specific, mesh-like, substructures within the central nervous system extracellular matrix. They form on specific subsets of neurons and are well-established as key regulators of plasticity. The appearance of PNNs coincides with the developmental transition of the brain from a more to less plastic state with numerous studies implicating PNNs in regulating this transition. Additionally, recent work has also linked PNNs to several neuropsychiatric and neurodevelopmental disorders. However, despite this growing interest in PNNs, the mechanisms by which they modulate neural functions are poorly understood. This limited mechanistic understanding of PNNs is derived from the fact that there are limited models, tools, or techniques that specifically target PNNs without disrupting the surrounding neural extracellular matrix. Our work, therefore, focuses on understanding how PNNs form on the surface of cells with the ultimate goal of developing models and tools to manipulate and disrupt PNNs specifically. Here, using a phosphatidylinositol specific phospholipase-C, we first demonstrate that PNN components are bound to the cell surface by a glycosylphosphatidylinositol-linked receptor protein. Furthermore, we demonstrate, through the exogenous addition of WT and a mutant variant of phoshpacan to primary cortical neurons, that contactin-1 is the glycosylphosphatidylinositol-linked protein critical for retaining nets to the cell surface. We believe the identification of contactin-1 as a key cell-surface protein for PNN structure is a very significant step forward in our understanding of the formation and structure of nets. It will offer new strategies to dissect the assembly of this specialized neural matrix.
Collapse
Affiliation(s)
- Ashis Sinha
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Gabrielle Nickerson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Samuel Bouyain
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA.
| | - Russell T Matthews
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA.
| |
Collapse
|
2
|
Sotogaku N, Nishi A. Chondroitin Sulfate E Regulates Neuronal Adhesion by Promoting Actin Polymerization Through Activation of the VEGF Receptor/Akt/GSK3β Pathway. Mol Neurobiol 2025:10.1007/s12035-025-04946-x. [PMID: 40295360 DOI: 10.1007/s12035-025-04946-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
Neuronal adhesion is regulated by interactions between neurons and the extracellular matrix and plays a critical role in neural development. Chondroitin sulfate proteoglycans (CSPGs), key structural components of the extracellular matrix in the nervous system, are involved in various processes including neuronal migration, neurite outgrowth, and axonal regeneration. The diverse functions of CSPGs are controlled by the sulfated structures of chondroitin sulfate (CS) polysaccharides. In this study, we found that CS-E, a highly sulfated CS polysaccharide, but not CS-A, CS-B, CS-C, or CS -D, induced the formation of neuronal aggregates in primary cortical cells with features of immature neurons. This effect is likely due to the unique properties of CS-E, which altered cell adhesion to the coated surface of culture coverslips, leading to cell detachment and subsequent aggregate formation. Pharmacological and phosphorylation analyses aimed at elucidating signaling cascades revealed that the VEGF receptor is a cell surface target of CS-E, and subsequent activation of the Akt/GSK3β pathway plays a crucial role in aggregate formation. The c-Raf/GSK3β pathway is also involved in CS-E-induced aggregate formation. Furthermore, actin polymerization and the organization of the F-actin cytoskeleton, which are mediated through activation of the Akt/GSK3β pathway, were required for CS-E-induced aggregate formation. In summary, CS-E regulates neuronal adhesion by activating the VEGF receptor/Akt/GSK3β pathway. The identification of signaling cascades involving CS-E may provide insights into the mechanisms underlying neuronal development.
Collapse
Affiliation(s)
- Naoki Sotogaku
- Laboratory of Functional Molecular Pharmaceutics, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, Aomori 030 - 0943, Japan.
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Fukuoka, 830 - 0011, Japan.
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Fukuoka, 830 - 0011, Japan
| |
Collapse
|
3
|
Hashimoto JG, Margolies N, Zhang X, Karpf J, Song Y, Gorham N, Davis BA, Zhang F, Linhardt RJ, Carbone L, Guizzetti M. Astrocyte Extracellular Matrix Modulates Neuronal Dendritic Development. Glia 2025. [PMID: 40192069 DOI: 10.1002/glia.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/25/2025]
Abstract
Major developmental events occurring in the hippocampus during the third trimester of human gestation and neonatally in altricial rodents include rapid and synchronized dendritic arborization and astrocyte proliferation and maturation. We tested the hypothesis that signals sent by developing astrocytes to developing neurons modulate dendritic development in vivo. First, we altered neuronal development by exposing neonatal (third trimester-equivalent) mice to ethanol, which increased dendritic arborization in hippocampal pyramidal neurons. We next assessed concurrent changes in the mouse astrocyte translatome by translating ribosomal affinity purification (TRAP)-seq. We followed up on ethanol-inhibition of astrocyte Chpf2 and Chsy1 gene translation because these genes encode biosynthetic enzymes of chondroitin sulfate glycosaminoglycan (CS-GAG) chains (extracellular matrix components that inhibit neuronal development and plasticity) and have not been explored before for their roles in dendritic arborization. We report that Chpf2 and Chsy1 are enriched in astrocytes, and their translation is inhibited by ethanol, which also reduces the levels of CS-GAGs measured by Liquid Chromatography/Mass Spectrometry. Finally, astrocyte-conditioned medium derived from Chfp2-silenced astrocytes increased neurite length and branching of hippocampal neurons in vitro, mechanistically linking changes in CS-GAG biosynthetic enzymes in astrocytes to altered neuronal development. These results demonstrate that CS-GAG biosynthetic enzymes in astrocytes regulate dendritic arborization in developing neurons and are involved in ethanol-induced altered neuronal development.
Collapse
Affiliation(s)
- Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Nicholas Margolies
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Xiaolu Zhang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Joshua Karpf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Yuefan Song
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Natalie Gorham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Brett A Davis
- Department of Medicine & Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Fuming Zhang
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Robert J Linhardt
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Lucia Carbone
- Department of Medicine & Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular and Genetics, Oregon Health & Science University, Portland, Oregon, USA
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
- Division of Genetics, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
4
|
Letko Khait N, Zuccaro S, Abdo D, Cui H, Siu R, Ho E, Morshead CM, Shoichet MS. Redesigned chondroitinase ABC degrades inhibitory chondroitin sulfate proteoglycans in vitro and in vivo in the stroke-injured rat brain. Biomaterials 2025; 314:122818. [PMID: 39260032 DOI: 10.1016/j.biomaterials.2024.122818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
Injuries to the central nervous system, such as stroke and traumatic spinal cord injury, result in an aggregate scar that both limits tissue degeneration and inhibits tissue regeneration. The aggregate scar includes chondroitin sulfate proteoglycans (CSPGs), which impede cell migration and axonal outgrowth. Chondroitinase ABC (ChASE) is a potent yet fragile enzyme that degrades CSPGs, and thus may enable tissue regeneration. ChASE37, with 37-point mutations to the native enzyme, has been shown to be more stable than ChASE, but its efficacy has never been tested. To answer this question, we investigated the efficacy of ChASE37 first in vitro using human cell-based assays and then in vivo in a rodent model of stroke. We demonstrated ChASE37 degradation of CSPGs in vitro and the consequent cell adhesion and axonal sprouting now possible using human induced pluripotent stem cell (hiPSC)-derived neurons. To enable prolonged release of ChASE37 to injured tissue, we expressed it as a fusion protein with a Src homology 3 (SH3) domain and modified an injectable, carboxymethylcellulose (CMC) hydrogel with SH3-binding peptides (CMC-bp) using inverse electron-demand Diels-Alder chemistry. We injected this affinity release CMC-bp/SH3-ChASE37 hydrogel epicortically to endothelin-1 stroke-injured rats and confirmed bioactivity via degradation of CSPGs and axonal sprouting in and around the lesion. With CSPG degradation shown both in vitro by greater cell interaction and in vivo with local delivery from a sustained release formulation, we lay the foundation to test the potential of ChASE37 and its delivery by local affinity release for tissue regeneration after stroke.
Collapse
Affiliation(s)
- Nitzan Letko Khait
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Sabrina Zuccaro
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Dhana Abdo
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada; Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada
| | - Hong Cui
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Ricky Siu
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada; Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada
| | - Eric Ho
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada; Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada
| | - Cindi M Morshead
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada; Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada; Department of Surgery, University of Toronto, 149 College Street, Toronto, ON, M5S 3E1, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada; Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON, M5S 3G9, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada.
| |
Collapse
|
5
|
Kirk RW, Sun L, Xiao R, Clark EA, Nelson S. Multiplexed CRISPRi Reveals a Transcriptional Switch Between KLF Activators and Repressors in the Maturing Neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636951. [PMID: 39975013 PMCID: PMC11839100 DOI: 10.1101/2025.02.07.636951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
A critical phase of mammalian brain development takes place after birth. Neurons of the mouse neocortex undergo dramatic changes in their morphology, physiology, and synaptic connections during the first postnatal month, while properties of immature neurons, such as the capacity for robust axon outgrowth, are lost. The genetic and epigenetic programs controlling prenatal development are well studied, but our understanding of the transcriptional mechanisms that regulate postnatal neuronal maturation is comparatively lacking. By integrating chromatin accessibility and gene expression data from two subtypes of neocortical pyramidal neurons in the neonatal and maturing brain, we predicted a role for the Krüppel-Like Factor (KLF) family of Transcription Factors in the developmental regulation of neonatally expressed genes. Using a multiplexed CRISPR Interference (CRISPRi) knockdown strategy, we found that a shift in expression from KLF activators (Klf6, Klf7) to repressors (Klf9, Klf13) during early postnatal development functions as a transcriptional 'switch' to first activate, then repress a set of shared targets with cytoskeletal functions including Tubb2b and Dpysl3. We demonstrate that this switch is buffered by redundancy between KLF paralogs, which our multiplexed CRISPRi strategy is equipped to overcome and study. Our results indicate that competition between activators and repressors within the KLF family regulates a conserved component of the postnatal maturation program that may underlie the loss of intrinsic axon growth in maturing neurons. This could facilitate the transition from axon growth to synaptic refinement required to stabilize mature circuits.
Collapse
Affiliation(s)
- Ryan W Kirk
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Liwei Sun
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Ruixuan Xiao
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Erin A Clark
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Sacha Nelson
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
6
|
Basabrain MS, Zaeneldin A, Bijle MN, Zhang C. Dental stem cell sphere formation and potential for neural regeneration: A scoping review. Heliyon 2024; 10:e40262. [PMID: 39619582 PMCID: PMC11605411 DOI: 10.1016/j.heliyon.2024.e40262] [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: 01/08/2024] [Revised: 10/26/2024] [Accepted: 11/07/2024] [Indexed: 01/31/2025] Open
Abstract
Background Dental stem cells with neurosphere-forming abilities are a promising cell source for the treatment of neural diseases and injuries. This scoping review aimed to systematically map the existing literature on dental sphere formation assays and their characteristics associated with neural regeneration potential. Methods The Web of Science, EMBASE, SCOPUS, and PubMed databases were systematically searched for in vitro, animal, and clinical studies and reviews focusing on stem cells isolated from the oral cavity, subsequently cultured as spheres with neural regeneration potential. Data were extracted and evidence was synthesized according to the predetermined variables in the registered protocol. Results A total of 35 articles (31 in vitro, 1 combined in vitro and in vivo, and 3 reviews) were included. The predominant method utilized for sphere formation was low-attachment culture. Spheres were characterized using assessment of neural marker expression via confocal microscopy, immunohistochemistry, RT-qPCR, or western blotting. Overall, the synthesized results indicate a lack of in vivo studies investigating the utility of dental neurospheres for neural regeneration, with dental pulp stem cells being the most investigated for their neural regenerative potential. Conclusion Dental stem cell spheres demonstrate significant potential for neural regeneration. Several assays and characterizations have been performed to characterized the mechanisms underlying dental sphere formation. Furthermore, in vivo studies are imperative to deduce the neural regenerative potential of stem cells in complex biological environments.
Collapse
Affiliation(s)
- Mohammed S. Basabrain
- Restorative Dental Sciences, Faculty of Dentistry, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmed Zaeneldin
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Mohammed Nadeem Bijle
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
7
|
Siwak M, Piotrzkowska D, Skrzypek M, Majsterek I. Effects of PEMF and LIPUS Therapy on the Expression of Genes Related to Peripheral Nerve Regeneration in Schwann Cells. Int J Mol Sci 2024; 25:12791. [PMID: 39684499 DOI: 10.3390/ijms252312791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Peripheral nerve regeneration remains a major challenge in neuroscience, despite advancements in understanding its mechanisms. Current treatments, including nerve transplantation and drug therapies, face limitations such as invasiveness and incomplete recovery of nerve function. Physical therapies, like pulsed electromagnetic fields (PEMF) and low-intensity ultrasound (LIPUS), are gaining attention for their potential to enhance regeneration. This study analyzes the effects of PEMF and LIPUS on gene expression in human primary Schwann cells, which are crucial for nerve myelination and repair. Key genes involved in neurotrophin signaling (NGF, BDNF), inflammation (IL-1β, IL-6, IL-10, TNF-α, TGF-β), and regeneration (CRYAB, CSPG, Ki67) were assessed. The results of this study reveal that combined PEMF and LIPUS therapies promote Schwann cell proliferation, reduce inflammation, and improve the regenerative environment, offering potential for optimizing these therapies for clinical use in regenerative medicine.
Collapse
Affiliation(s)
- Mateusz Siwak
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Danuta Piotrzkowska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Maciej Skrzypek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| |
Collapse
|
8
|
Sinha A, Nickerson G, Bouyain S, Matthews RT. Contactin-1 is a critical neuronal cell surface receptor for perineuronal net structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622114. [PMID: 39605332 PMCID: PMC11601535 DOI: 10.1101/2024.11.05.622114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Perineuronal nets (PNNs), are neuron-specific substructures within the neural extracellular matrix (ECM). These reticular structures form on a very small subset of neurons in the central nervous system (CNS) and yet have a profound impact in regulating neuronal development and physiology. PNNs are well-established as key regulators of plasticity in the CNS. Their appearance coincides with the developmental transition of the brain more to less plastic state. And, importantly, numerous studies have demonstrated that indeed PNNs play a primary role in regulating this transition. There is, however, a growing literature implicating PNNs in numerous roles in neural physiology beyond their role in regulating developmental plasticity. Accordingly, numerous studies have shown PNNs are altered in a variety of neurological and neuropsychiatric diseases, linking them to these conditions. Despite the growing interest in PNNs, the mechanisms by which they modulate neural functions are poorly understood. We believe the limited mechanistic understanding of PNNs is derived from the fact that there are limited models, tools or techniques that specifically target PNNs in a cell-autonomous manner and without also disrupting the surrounding neural ECM. These limitations are primarily due to our incomplete understanding of PNN composition and structure. In particular, there is little understanding of the neuronal cell surface receptors that nucleate these structures on subset of neurons on which they form in the CNS. Therefore, the main focus our work is to identify the neuronal cell surface proteins critical for PNN formation and structure. In our previous studies we demonstrated PNN components are immobilized on the neuronal surface by two distinct mechanisms, one dependent on the hyaluronan backbone of PNNs and the other mediated by a complex formed by receptor protein tyrosine phosphatase zeta (RPTPζ) and tenascin-R (Tnr). Here we first demonstrate that the Tnr-RPTPζ complex in PNNs is bound to the cell surface by a glycosylphosphatidylinositol (GPI)-linked receptor protein. Using a biochemical and structural approach we demonstrate the GPI-linked protein critical for binding the Tnr-RPTPζ complex in PNNs is contactin-1 (Cntn1). We further show the binding of this complex in PNNs by Cntn1 is critical for PNN structure. We believe identification of CNTN1 as a key cell-surface protein for PNN structure is a very significant step forward in our understanding of PNN formation and structure and will offer new strategies and targets to manipulate PNNs and better understand their function.
Collapse
Affiliation(s)
- Ashis Sinha
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York 13210
| | - Gabrielle Nickerson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York 13210
| | - Samuel Bouyain
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri 64110
| | - Russell T. Matthews
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
9
|
Brown RI, Barber HM, Kucenas S. Satellite glial cell manipulation prior to axotomy enhances developing dorsal root ganglion central branch regrowth into the spinal cord. Glia 2024; 72:1766-1784. [PMID: 39141572 PMCID: PMC11325082 DOI: 10.1002/glia.24581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 08/16/2024]
Abstract
The central and peripheral nervous systems (CNS and PNS, respectively) exhibit remarkable diversity in the capacity to regenerate following neuronal injury with PNS injuries being much more likely to regenerate than those that occur in the CNS. Glial responses to damage greatly influence the likelihood of regeneration by either promoting or inhibiting axonal regrowth over time. However, despite our understanding of how some glial lineages participate in nerve degeneration and regeneration, less is known about the contributions of peripheral satellite glial cells (SGC) to regeneration failure following central axon branch injury of dorsal root ganglia (DRG) sensory neurons. Here, using in vivo, time-lapse imaging in larval zebrafish coupled with laser axotomy, we investigate the role of SGCs in axonal regeneration. In our studies we show that SGCs respond to injury by relocating their nuclei to the injury site during the same period that DRG neurons produce new central branch neurites. Laser ablation of SGCs prior to axon injury results in more neurite growth attempts and ultimately a higher rate of successful central axon regrowth, implicating SGCs as inhibitors of regeneration. We also demonstrate that this SGC response is mediated in part by ErbB signaling, as chemical inhibition of this receptor results in reduced SGC motility and enhanced central axon regrowth. These findings provide new insights into SGC-neuron interactions under injury conditions and how these interactions influence nervous system repair.
Collapse
Affiliation(s)
- Robin I Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Heather M Barber
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Cell & Developmental Biology Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
10
|
Hashimoto JG, Margolies N, Zhang X, Karpf J, Song Y, Davis BA, Zhang F, Linhardt RJ, Carbone L, Guizzetti M. Astrocyte extracellular matrix modulates neuronal dendritic development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606424. [PMID: 39211148 PMCID: PMC11361265 DOI: 10.1101/2024.08.06.606424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Major developmental events occurring in the hippocampus during the third trimester of human gestation and neonatally in altricial rodents include rapid and synchronized dendritic arborization and astrocyte proliferation and maturation. We tested the hypothesis that signals sent by developing astrocytes to developing neurons modulate dendritic development in vivo. We altered neuronal development by neonatal (third trimester-equivalent) ethanol exposure in mice; this treatment increased dendritic arborization in hippocampal pyramidal neurons. We next assessed concurrent changes in the mouse astrocyte translatome by translating ribosomal affinity purification (TRAP)-seq. We followed up on ethanol-inhibition of astrocyte Chpf2 and Chsy1 gene translation because these genes encode for biosynthetic enzymes of chondroitin sulfate glycosaminoglycan (CS-GAG) chains (extracellular matrix components that inhibit neuronal development and plasticity) and have not been explored before for their roles in dendritic arborization. We report that Chpf2 and Chsy1 are enriched in astrocytes and their translation is inhibited by ethanol, which also reduces the levels of CS-GAGs measured by Liquid Chromatography/Mass Spectrometry. Finally, astrocyte-conditioned medium derived from Chfp2-silenced astrocytes increased neurite branching of hippocampal neurons in vitro. These results demonstrate that CS-GAG biosynthetic enzymes in astrocytes regulates dendritic arborization in developing neurons.
Collapse
Affiliation(s)
- Joel G. Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- VA Portland Health Care System, Portland, OR
| | - Nicholas Margolies
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- VA Portland Health Care System, Portland, OR
| | - Xiaolu Zhang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- VA Portland Health Care System, Portland, OR
| | - Joshua Karpf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | - Yuefan Song
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Brett A. Davis
- Department of Medicine & Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Fuming Zhang
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Robert J. Linhardt
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Lucia Carbone
- Department of Medicine & Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
- Department of Molecular and Genetics, Oregon Health & Science University, Portland, OR
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR
- Division of Genetics, Oregon National Primate Research Center, Beaverton, OR
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- VA Portland Health Care System, Portland, OR
| |
Collapse
|
11
|
Sethi MK, Maccioni R, Hogan JD, Kawamura T, Repunte-Canonigo V, Chen J, Zaia J, Sanna PP. Comprehensive Glycomic and Proteomic Analysis of Mouse Striatum and Lateral Hypothalamus Following Repeated Exposures to Cocaine or Methamphetamine. Mol Cell Proteomics 2024; 23:100803. [PMID: 38880242 PMCID: PMC11324981 DOI: 10.1016/j.mcpro.2024.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024] Open
Abstract
Substance use disorder is a major concern, with few therapeutic options. Heparan sulfate (HS) and chondroitin sulfate (CS) interact with a plethora of growth factors and their receptors and have profound effects on cellular signaling. Thus, targeting these dynamic interactions might represent a potential novel therapeutic modality. In the present study, we performed mass spectrometry-based glycomic and proteomic analysis to understand the effects of cocaine and methamphetamine (METH) on HS, CS, and the proteome of two brain regions critically involved in drug addiction: the lateral hypothalamus and the striatum. We observed that cocaine and METH significantly alter HS and CS abundances as well as sulfate contents and composition. In particular, repeated METH or cocaine treatments reduced CS 4-O-sulfation and increased CS 6-O-sulfation. Since C4S and C6S exercise differential effects on axon growth, regeneration, and plasticity, these changes likely contribute to drug-induced neural plasticity in these brain regions. Notably, we observed that restoring these alterations by increasing CS 4-0 levels in the lateral hypothalamus by adeno-associated virus delivery of an shRNA to arylsulfatase B (N-acetylgalactosamine-4-sulfatase) ameliorated anxiety and prevented the expression of preference for cocaine in a novelty induced conditioned place preference test during cocaine withdrawal. Finally, proteomics analyses revealed a number of aberrant proteins in METH- and cocaine-treated versus saline-treated mice, including myelin proteolipid protein, calcium/calmodulin-dependent protein kinase type II subunit alpha, synapsin-2, tenascin-R, calnexin, annexin A7, hepatoma-derived growth factor, neurocan, and CSPG5, and oxidative phosphorylation among the top perturbed pathway. Taken together, these data support the role of HS, CS, and associated proteins in stimulants abuse and suggest that manipulation of HSPGs can represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Manveen K Sethi
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Riccardo Maccioni
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - John D Hogan
- Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Tomoya Kawamura
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Jihuan Chen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Joseph Zaia
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University School of Medicine, Boston, Massachusetts, USA; Bioinformatics Program, Boston University, Boston, Massachusetts, USA.
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA.
| |
Collapse
|
12
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
13
|
Gómez-Oliva R, Geribaldi-Doldán N, Domínguez-García S, Pardillo-Díaz R, Martínez-Ortega S, Oliva-Montero JM, Pérez-García P, García-Cózar FJ, Muñoz-Miranda JP, Sánchez-Gomar I, Nunez-Abades P, Castro C. Targeting epidermal growth factor receptor to recruit newly generated neuroblasts in cortical brain injuries. J Transl Med 2023; 21:867. [PMID: 38037126 PMCID: PMC10687845 DOI: 10.1186/s12967-023-04707-1] [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: 02/03/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Neurogenesis is stimulated in the subventricular zone (SVZ) of mice with cortical brain injuries. In most of these injuries, newly generated neuroblasts attempt to migrate toward the injury, accumulating within the corpus callosum not reaching the perilesional area. METHODS We use a murine model of mechanical cortical brain injury, in which we perform unilateral cortical injuries in the primary motor cortex of adult male mice. We study neurogenesis in the SVZ and perilesional area at 7 and 14 dpi as well as the expression and concentration of the signaling molecule transforming growth factor alpha (TGF-α) and its receptor the epidermal growth factor (EGFR). We use the EGFR inhibitor Afatinib to promote neurogenesis in brain injuries. RESULTS We show that microglial cells that emerge within the injured area and the SVZ in response to the injury express high levels of TGF-α leading to elevated concentrations of TGF-α in the cerebrospinal fluid. Thus, the number of neuroblasts in the SVZ increases in response to the injury, a large number of these neuroblasts remain immature and proliferate expressing the epidermal growth factor receptor (EGFR) and the proliferation marker Ki67. Restraining TGF-α release with a classical protein kinase C inhibitor reduces the number of these proliferative EGFR+ immature neuroblasts in the SVZ. In accordance, the inhibition of the TGF-α receptor, EGFR promotes migration of neuroblasts toward the injury leading to an elevated number of neuroblasts within the perilesional area. CONCLUSIONS Our results indicate that in response to an injury, microglial cells activated within the injury and the SVZ release TGF-α, activating the EGFR present in the neuroblasts membrane inducing their proliferation, delaying maturation and negatively regulating migration. The inactivation of this signaling pathway stimulates neuroblast migration toward the injury and enhances the quantity of neuroblasts within the injured area. These results suggest that these proteins may be used as target molecules to regenerate brain injuries.
Collapse
Affiliation(s)
- Ricardo Gómez-Oliva
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Noelia Geribaldi-Doldán
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Ricardo Pardillo-Díaz
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Sergio Martínez-Ortega
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - José M Oliva-Montero
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Patricia Pérez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Francisco J García-Cózar
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Área de Inmunología, Universidad de Cádiz, Cádiz, Spain
| | - Juan P Muñoz-Miranda
- Servicios Centrales de Investigación Biomédica, Universidad de Cádiz, Cádiz, Spain
| | - Ismael Sánchez-Gomar
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain
- Departamento de Fisiología, Universidad de Sevilla, Sevilla, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.
- Instituto de Investigación e Innovación Biomédica de Cádiz, Cádiz, Spain.
| |
Collapse
|
14
|
Chemistry and Function of Glycosaminoglycans in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:117-162. [DOI: 10.1007/978-3-031-12390-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
15
|
Abad-Rodríguez J, Brocca ME, Higuero AM. Glycans and Carbohydrate-Binding/Transforming Proteins in Axon Physiology. ADVANCES IN NEUROBIOLOGY 2023; 29:185-217. [PMID: 36255676 DOI: 10.1007/978-3-031-12390-0_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The mature nervous system relies on the polarized morphology of neurons for a directed flow of information. These highly polarized cells use their somatodendritic domain to receive and integrate input signals while the axon is responsible for the propagation and transmission of the output signal. However, the axon must perform different functions throughout development before being fully functional for the transmission of information in the form of electrical signals. During the development of the nervous system, axons perform environmental sensing functions, which allow them to navigate through other regions until a final target is reached. Some axons must also establish a regulated contact with other cells before reaching maturity, such as with myelinating glial cells in the case of myelinated axons. Mature axons must then acquire the structural and functional characteristics that allow them to perform their role as part of the information processing and transmitting unit that is the neuron. Finally, in the event of an injury to the nervous system, damaged axons must try to reacquire some of their immature characteristics in a regeneration attempt, which is mostly successful in the PNS but fails in the CNS. Throughout all these steps, glycans perform functions of the outermost importance. Glycans expressed by the axon, as well as by their surrounding environment and contacting cells, encode key information, which is fine-tuned by glycan modifying enzymes and decoded by glycan binding proteins so that the development, guidance, myelination, and electrical transmission functions can be reliably performed. In this chapter, we will provide illustrative examples of how glycans and their binding/transforming proteins code and decode instructive information necessary for fundamental processes in axon physiology.
Collapse
Affiliation(s)
- José Abad-Rodríguez
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain.
| | - María Elvira Brocca
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Alonso Miguel Higuero
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| |
Collapse
|
16
|
Wang Q, Chi L. The Alterations and Roles of Glycosaminoglycans in Human Diseases. Polymers (Basel) 2022; 14:polym14225014. [PMID: 36433141 PMCID: PMC9694910 DOI: 10.3390/polym14225014] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Glycosaminoglycans (GAGs) are a heterogeneous family of linear polysaccharides which are composed of a repeating disaccharide unit. They are also linked to core proteins to form proteoglycans (PGs). GAGs/PGs are major components of the cell surface and the extracellular matrix (ECM), and they display critical roles in development, normal function, and damage response in the body. Some properties (such as expression quantity, molecular weight, and sulfation pattern) of GAGs may be altered under pathological conditions. Due to the close connection between these properties and the function of GAGs/PGs, the alterations are often associated with enormous changes in the physiological/pathological status of cells and organs. Therefore, these GAGs/PGs may serve as marker molecules of disease. This review aimed to investigate the structural alterations and roles of GAGs/PGs in a range of diseases, such as atherosclerosis, cancer, diabetes, neurodegenerative disease, and virus infection. It is hoped to provide a reference for disease diagnosis, monitoring, prognosis, and drug development.
Collapse
|
17
|
Tewari BP, Chaunsali L, Prim CE, Sontheimer H. A glial perspective on the extracellular matrix and perineuronal net remodeling in the central nervous system. Front Cell Neurosci 2022; 16:1022754. [PMID: 36339816 PMCID: PMC9630365 DOI: 10.3389/fncel.2022.1022754] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
A structural scaffold embedding brain cells and vasculature is known as extracellular matrix (ECM). The physical appearance of ECM in the central nervous system (CNS) ranges from a diffused, homogeneous, amorphous, and nearly omnipresent matrix to highly organized distinct morphologies such as basement membranes and perineuronal nets (PNNs). ECM changes its composition and organization during development, adulthood, aging, and in several CNS pathologies. This spatiotemporal dynamic nature of the ECM and PNNs brings a unique versatility to their functions spanning from neurogenesis, cell migration and differentiation, axonal growth, and pathfinding cues, etc., in the developing brain, to stabilizing synapses, neuromodulation, and being an active partner of tetrapartite synapses in the adult brain. The malleability of ECM and PNNs is governed by both intrinsic and extrinsic factors. Glial cells are among the major extrinsic factors that facilitate the remodeling of ECM and PNN, thereby acting as key regulators of diverse functions of ECM and PNN in health and diseases. In this review, we discuss recent advances in our understanding of PNNs and how glial cells are central to ECM and PNN remodeling in normal and pathological states of the CNS.
Collapse
|
18
|
Couto MR, Rodrigues JL, Rodrigues LR. Heterologous production of chondroitin. BIOTECHNOLOGY REPORTS 2022; 33:e00710. [PMID: 35242620 PMCID: PMC8858990 DOI: 10.1016/j.btre.2022.e00710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/17/2022] [Accepted: 02/08/2022] [Indexed: 01/01/2023]
Abstract
Chondroitin sulfate (CS) is a glycosaminoglycan with a growing variety of applications. CS can be produced from microbial fermentation of native or engineered strains. Synthetic biology tools are being used to improve CS yields in different hosts. Integrated polymerization and sulfation can generate cost-effective CS.
Chondroitin sulfate (CS) is a glycosaminoglycan with a broad range of applications being a popular dietary supplement for osteoarthritis. Usually, CS is extracted from animal sources. However, the known risks of animal products use have been driving the search for alternative methods and sources to obtain this compound. Several pathogenic bacteria naturally produce chondroitin-like polysaccharides through well-known pathways and, therefore, have been the basis for numerous studies that aim to produce chondroitin using non-pathogenic hosts. However, the yields obtained are not enough to meet the high demand for this glycosaminoglycan. Metabolic engineering strategies have been used to construct improved heterologous hosts. The identification of metabolic bottlenecks and regulation points, and the screening for efficient enzymes are key points for constructing microbial cell factories with improved chondroitin yields to achieve industrial CS production. The recent advances on enzymatic and microbial strategies to produce non-animal chondroitin are herein reviewed. Challenges and prospects for future research are also discussed.
Collapse
Affiliation(s)
- Márcia R. Couto
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
| | - Joana L. Rodrigues
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
- Corresponding author.
| | - Lígia R. Rodrigues
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
19
|
Delalande JM, Nagy N, McCann CJ, Natarajan D, Cooper JE, Carreno G, Dora D, Campbell A, Laurent N, Kemos P, Thomas S, Alby C, Attié-Bitach T, Lyonnet S, Logan MP, Goldstein AM, Davey MG, Hofstra RMW, Thapar N, Burns AJ. TALPID3/KIAA0586 Regulates Multiple Aspects of Neuromuscular Patterning During Gastrointestinal Development in Animal Models and Human. Front Mol Neurosci 2022; 14:757646. [PMID: 35002618 PMCID: PMC8733242 DOI: 10.3389/fnmol.2021.757646] [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: 08/12/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022] Open
Abstract
TALPID3/KIAA0586 is an evolutionary conserved protein, which plays an essential role in protein trafficking. Its role during gastrointestinal (GI) and enteric nervous system (ENS) development has not been studied previously. Here, we analyzed chicken, mouse and human embryonic GI tissues with TALPID3 mutations. The GI tract of TALPID3 chicken embryos was shortened and malformed. Histologically, the gut smooth muscle was mispatterned and enteric neural crest cells were scattered throughout the gut wall. Analysis of the Hedgehog pathway and gut extracellular matrix provided causative reasons for these defects. Interestingly, chicken intra-species grafting experiments and a conditional knockout mouse model showed that ENS formation did not require TALPID3, but was dependent on correct environmental cues. Surprisingly, the lack of TALPID3 in enteric neural crest cells (ENCC) affected smooth muscle and epithelial development in a non-cell-autonomous manner. Analysis of human gut fetal tissues with a KIAA0586 mutation showed strikingly similar findings compared to the animal models demonstrating conservation of TALPID3 and its necessary role in human GI tract development and patterning.
Collapse
Affiliation(s)
- Jean Marie Delalande
- Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Dipa Natarajan
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Julie E Cooper
- Developmental Biology and Cancer Program, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Gabriela Carreno
- Developmental Biology and Cancer Program, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Alison Campbell
- Department of Paediatric Surgery, Christchurch Hospital, Christchurch, New Zealand
| | - Nicole Laurent
- Génétique et Anomalies du Développement, Université de Bourgogne, Service d'Anatomie Pathologique, Dijon, France
| | - Polychronis Kemos
- Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sophie Thomas
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France
| | - Caroline Alby
- Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Tania Attié-Bitach
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France.,Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France.,Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stanislas Lyonnet
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France.,Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France.,Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Malcolm P Logan
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Megan G Davey
- Division of Developmental Biology, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Division of Neurogastroenterology and Motility, Department of Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom.,Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Inc., Cambridge, MA, United States
| |
Collapse
|
20
|
Mizumoto S, Yamada S. An Overview of in vivo Functions of Chondroitin Sulfate and Dermatan Sulfate Revealed by Their Deficient Mice. Front Cell Dev Biol 2021; 9:764781. [PMID: 34901009 PMCID: PMC8652114 DOI: 10.3389/fcell.2021.764781] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Chondroitin sulfate (CS), dermatan sulfate (DS) and heparan sulfate (HS) are covalently attached to specific core proteins to form proteoglycans in their biosynthetic pathways. They are constructed through the stepwise addition of respective monosaccharides by various glycosyltransferases and maturated by epimerases as well as sulfotransferases. Structural diversities of CS/DS and HS are essential for their various biological activities including cell signaling, cell proliferation, tissue morphogenesis, and interactions with a variety of growth factors as well as cytokines. Studies using mice deficient in enzymes responsible for the biosynthesis of the CS/DS and HS chains of proteoglycans have demonstrated their essential functions. Chondroitin synthase 1-deficient mice are viable, but exhibit chondrodysplasia, progression of the bifurcation of digits, delayed endochondral ossification, and reduced bone density. DS-epimerase 1-deficient mice show thicker collagen fibrils in the dermis and hypodermis, and spina bifida. These observations suggest that CS/DS are essential for skeletal development as well as the assembly of collagen fibrils in the skin, and that their respective knockout mice can be utilized as models for human genetic disorders with mutations in chondroitin synthase 1 and DS-epimerase 1. This review provides a comprehensive overview of mice deficient in CS/DS biosyntheses.
Collapse
Affiliation(s)
- Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| |
Collapse
|
21
|
Malekmohammadi S, Sedghi Aminabad N, Sabzi A, Zarebkohan A, Razavi M, Vosough M, Bodaghi M, Maleki H. Smart and Biomimetic 3D and 4D Printed Composite Hydrogels: Opportunities for Different Biomedical Applications. Biomedicines 2021; 9:1537. [PMID: 34829766 PMCID: PMC8615087 DOI: 10.3390/biomedicines9111537] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/10/2021] [Accepted: 10/16/2021] [Indexed: 12/17/2022] Open
Abstract
In recent years, smart/stimuli-responsive hydrogels have drawn tremendous attention for their varied applications, mainly in the biomedical field. These hydrogels are derived from different natural and synthetic polymers but are also composite with various organic and nano-organic fillers. The basic functions of smart hydrogels rely on their ability to change behavior; functions include mechanical, swelling, shaping, hydrophilicity, and bioactivity in response to external stimuli such as temperature, pH, magnetic field, electromagnetic radiation, and biological molecules. Depending on the final applications, smart hydrogels can be processed in different geometries and modalities to meet the complicated situations in biological media, namely, injectable hydrogels (following the sol-gel transition), colloidal nano and microgels, and three dimensional (3D) printed gel constructs. In recent decades smart hydrogels have opened a new horizon for scientists to fabricate biomimetic customized biomaterials for tissue engineering, cancer therapy, wound dressing, soft robotic actuators, and controlled release of bioactive substances/drugs. Remarkably, 4D bioprinting, a newly emerged technology/concept, aims to rationally design 3D patterned biological matrices from synthesized hydrogel-based inks with the ability to change structure under stimuli. This technology has enlarged the applicability of engineered smart hydrogels and hydrogel composites in biomedical fields. This paper aims to review stimuli-responsive hydrogels according to the kinds of external changes and t recent applications in biomedical and 4D bioprinting.
Collapse
Affiliation(s)
- Samira Malekmohammadi
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK;
- Department of Regenerative Medicine, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
- Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran;
| | - Negar Sedghi Aminabad
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran; (N.S.A.); (A.S.)
| | - Amin Sabzi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran; (N.S.A.); (A.S.)
| | - Amir Zarebkohan
- Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran;
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran; (N.S.A.); (A.S.)
| | - Mehdi Razavi
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| | - Massoud Vosough
- Department of Regenerative Medicine, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Mahdi Bodaghi
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK;
| | - Hajar Maleki
- Department of Chemistry, Institute of Inorganic Chemistry, University of Cologne, 50939 Cologne, Germany
| |
Collapse
|
22
|
Abstract
Perineuronal nets (PNNs) are condensed extracellular matrix (ECM) assemblies of
polyanionic chondroitin sulfate proteoglycans, hyaluronan, and tenascins that
primarily wrap around GABAergic parvalbumin (PV) interneurons. During
development, PNN formation terminates the critical period of neuroplasticity, a
process that can be reversed by experimental disruption of PNNs. Perineuronal
nets also regulate the intrinsic properties of the enclosed PV neurons thereby
maintaining their inhibitory activity. Recent studies have implicated PNNs in
central nervous system diseases as well as PV neuron dysfunction; consequently,
they have further been associated with altered inhibition, particularly in the
genesis of epilepsy. A wide range of seizure presentations in human and rodent
models exhibit ECM remodeling with PNN disruption due to elevated protease
activity. Inhibition of PNN proteolysis reduces seizure activity suggesting that
PNN degrading enzymes may be potential novel therapeutic targets.
Collapse
Affiliation(s)
- Lata Chaunsali
- School of Neuroscience Graduate Program, Virginia Tech, Blacksburg, VA, USA.,Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Bhanu P Tewari
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| |
Collapse
|
23
|
Rial-Hermida MI, Rey-Rico A, Blanco-Fernandez B, Carballo-Pedrares N, Byrne EM, Mano JF. Recent Progress on Polysaccharide-Based Hydrogels for Controlled Delivery of Therapeutic Biomolecules. ACS Biomater Sci Eng 2021; 7:4102-4127. [PMID: 34137581 PMCID: PMC8919265 DOI: 10.1021/acsbiomaterials.0c01784] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 06/02/2021] [Indexed: 12/24/2022]
Abstract
A plethora of applications using polysaccharides have been developed in recent years due to their availability as well as their frequent nontoxicity and biodegradability. These polymers are usually obtained from renewable sources or are byproducts of industrial processes, thus, their use is collaborative in waste management and shows promise for an enhanced sustainable circular economy. Regarding the development of novel delivery systems for biotherapeutics, the potential of polysaccharides is attractive for the previously mentioned properties and also for the possibility of chemical modification of their structures, their ability to form matrixes of diverse architectures and mechanical properties, as well as for their ability to maintain bioactivity following incorporation of the biomolecules into the matrix. Biotherapeutics, such as proteins, growth factors, gene vectors, enzymes, hormones, DNA/RNA, and antibodies are currently in use as major therapeutics in a wide range of pathologies. In the present review, we summarize recent progress in the development of polysaccharide-based hydrogels of diverse nature, alone or in combination with other polymers or drug delivery systems, which have been implemented in the delivery of biotherapeutics in the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- M. Isabel Rial-Hermida
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| | - Ana Rey-Rico
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Barbara Blanco-Fernandez
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology, 08028 Barcelona, Spain
- CIBER
en Bioingeniería, Biomateriales y
Nanomedicina, CIBER-BBN, 28029 Madrid, Spain
| | - Natalia Carballo-Pedrares
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Eimear M. Byrne
- Wellcome-Wolfson
Institute For Experimental Medicine, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - João F. Mano
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| |
Collapse
|
24
|
Nishimura K, Takata K. Combination of Drugs and Cell Transplantation: More Beneficial Stem Cell-Based Regenerative Therapies Targeting Neurological Disorders. Int J Mol Sci 2021; 22:ijms22169047. [PMID: 34445753 PMCID: PMC8396512 DOI: 10.3390/ijms22169047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/02/2023] Open
Abstract
Cell transplantation therapy using pluripotent/multipotent stem cells has gained attention as a novel therapeutic strategy for treating neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, ischemic stroke, and spinal cord injury. To fully realize the potential of cell transplantation therapy, new therapeutic options that increase cell engraftments must be developed, either through modifications to the grafted cells themselves or through changes in the microenvironment surrounding the grafted region. Together these developments could potentially restore lost neuronal function by better supporting grafted cells. In addition, drug administration can improve the outcome of cell transplantation therapy through better accessibility and delivery to the target region following cell transplantation. Here we introduce examples of drug repurposing approaches for more successful transplantation therapies based on preclinical experiments with clinically approved drugs. Drug repurposing is an advantageous drug development strategy because drugs that have already been clinically approved can be repurposed to treat other diseases faster and at lower cost. Therefore, drug repurposing is a reasonable approach to enhance the outcomes of cell transplantation therapies for neurological diseases. Ideal repurposing candidates would result in more efficient cell transplantation therapies and provide a new and beneficial therapeutic combination.
Collapse
|
25
|
Tran AP, Warren PM, Silver J. New insights into glial scar formation after spinal cord injury. Cell Tissue Res 2021; 387:319-336. [PMID: 34076775 PMCID: PMC8975767 DOI: 10.1007/s00441-021-03477-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
Severe spinal cord injury causes permanent loss of function and sensation throughout the body. The trauma causes a multifaceted torrent of pathophysiological processes which ultimately act to form a complex structure, permanently remodeling the cellular architecture and extracellular matrix. This structure is traditionally termed the glial/fibrotic scar. Similar cellular formations occur following stroke, infection, and neurodegenerative diseases of the central nervous system (CNS) signifying their fundamental importance to preservation of function. It is increasingly recognized that the scar performs multiple roles affecting recovery following traumatic injury. Innovative research into the properties of this structure is imperative to the development of treatment strategies to recover motor function and sensation following CNS trauma. In this review, we summarize how the regeneration potential of the CNS alters across phyla and age through formation of scar-like structures. We describe how new insights from next-generation sequencing technologies have yielded a more complex portrait of the molecular mechanisms governing the astrocyte, microglial, and neuronal responses to injury and development, especially of the glial component of the scar. Finally, we discuss possible combinatorial therapeutic approaches centering on scar modulation to restore function after severe CNS injury.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Philippa Mary Warren
- Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, UK
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
26
|
Pantazopoulos H, Katsel P, Haroutunian V, Chelini G, Klengel T, Berretta S. Molecular signature of extracellular matrix pathology in schizophrenia. Eur J Neurosci 2021; 53:3960-3987. [PMID: 33070392 PMCID: PMC8359380 DOI: 10.1111/ejn.15009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Growing evidence points to a critical involvement of the extracellular matrix (ECM) in the pathophysiology of schizophrenia (SZ). Decreases of perineuronal nets (PNNs) and altered expression of chondroitin sulphate proteoglycans (CSPGs) in glial cells have been identified in several brain regions. GWAS data have identified several SZ vulnerability variants of genes encoding for ECM molecules. Given the potential relevance of ECM functions to the pathophysiology of this disorder, it is necessary to understand the extent of ECM changes across brain regions, their region- and sex-specificity and which ECM components contribute to these changes. We tested the hypothesis that the expression of genes encoding for ECM molecules may be broadly disrupted in SZ across several cortical and subcortical brain regions and include key ECM components as well as factors such as ECM posttranslational modifications and regulator factors. Gene expression profiling of 14 neocortical brain regions, caudate, putamen and hippocampus from control subjects (n = 14/region) and subjects with SZ (n = 16/region) was conducted using Affymetrix microarray analysis. Analysis across brain regions revealed widespread dysregulation of ECM gene expression in cortical and subcortical brain regions in SZ, impacting several ECM functional key components. SRGN, CD44, ADAMTS1, ADAM10, BCAN, NCAN and SEMA4G showed some of the most robust changes. Region-, sex- and age-specific gene expression patterns and correlation with cognitive scores were also detected. Taken together, these findings contribute to emerging evidence for large-scale ECM dysregulation in SZ and point to molecular pathways involved in PNN decreases, glial cell dysfunction and cognitive impairment in SZ.
Collapse
Affiliation(s)
- Harry Pantazopoulos
- Department of Neurobiology and Anatomical SciencesUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Pavel Katsel
- Department of PsychiatryThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of NeuroscienceThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mental Illness Research Education ClinicalCenters of Excellence (MIRECC)JJ Peters VA Medical CenterBronxNYUSA
| | - Vahram Haroutunian
- Department of PsychiatryThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of NeuroscienceThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mental Illness Research Education ClinicalCenters of Excellence (MIRECC)JJ Peters VA Medical CenterBronxNYUSA
| | - Gabriele Chelini
- Translational Neuroscience LaboratoryMclean HospitalBelmontMAUSA
- Department of PsychiatryHarvard Medical SchoolBostonMAUSA
| | - Torsten Klengel
- Department of PsychiatryHarvard Medical SchoolBostonMAUSA
- Translational Molecular Genomics LaboratoryMclean HospitalBelmontMAUSA
- Department of PsychiatryUniversity Medical Center GöttingenGöttingenGermany
| | - Sabina Berretta
- Translational Neuroscience LaboratoryMclean HospitalBelmontMAUSA
- Department of PsychiatryHarvard Medical SchoolBostonMAUSA
- Program in NeuroscienceHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
27
|
Melrose J, Hayes AJ, Bix G. The CNS/PNS Extracellular Matrix Provides Instructive Guidance Cues to Neural Cells and Neuroregulatory Proteins in Neural Development and Repair. Int J Mol Sci 2021; 22:5583. [PMID: 34070424 PMCID: PMC8197505 DOI: 10.3390/ijms22115583] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The extracellular matrix of the PNS/CNS is unusual in that it is dominated by glycosaminoglycans, especially hyaluronan, whose space filling and hydrating properties make essential contributions to the functional properties of this tissue. Hyaluronan has a relatively simple structure but its space-filling properties ensure micro-compartments are maintained in the brain ultrastructure, ensuring ionic niches and gradients are maintained for optimal cellular function. Hyaluronan has cell-instructive, anti-inflammatory properties and forms macro-molecular aggregates with the lectican CS-proteoglycans, forming dense protective perineuronal net structures that provide neural and synaptic plasticity and support cognitive learning. AIMS To highlight the central nervous system/peripheral nervous system (CNS/PNS) and its diverse extracellular and cell-associated proteoglycans that have cell-instructive properties regulating neural repair processes and functional recovery through interactions with cell adhesive molecules, receptors and neuroregulatory proteins. Despite a general lack of stabilising fibrillar collagenous and elastic structures in the CNS/PNS, a sophisticated dynamic extracellular matrix is nevertheless important in tissue form and function. CONCLUSIONS This review provides examples of the sophistication of the CNS/PNS extracellular matrix, showing how it maintains homeostasis and regulates neural repair and regeneration.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern, The University of Sydney, Sydney, NSW 2052, Australia
- Faculty of Medicine and Health, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK;
| | - Gregory Bix
- Clinical Neuroscience Research Center, Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| |
Collapse
|
28
|
Baker L, Tar M, Kramer AH, Villegas GA, Charafeddine RA, Vafaeva O, Nacharaju P, Friedman J, Davies KP, Sharp DJ. Fidgetin-like 2 negatively regulates axonal growth and can be targeted to promote functional nerve regeneration. JCI Insight 2021; 6:138484. [PMID: 33872220 PMCID: PMC8262307 DOI: 10.1172/jci.insight.138484] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
The microtubule (MT) cytoskeleton plays a critical role in axon growth and guidance. Here, we identify the MT-severing enzyme fidgetin-like 2 (FL2) as a negative regulator of axon regeneration and a therapeutic target for promoting nerve regeneration after injury. Genetic knockout of FL2 in cultured adult dorsal root ganglion neurons resulted in longer axons and attenuated growth cone retraction in response to inhibitory molecules. Given the axonal growth-promoting effects of FL2 depletion in vitro, we tested whether FL2 could be targeted to promote regeneration in a rodent model of cavernous nerve (CN) injury. The CNs are parasympathetic nerves that regulate blood flow to the penis, which are commonly damaged during radical prostatectomy (RP), resulting in erectile dysfunction (ED). Application of FL2-siRNA after CN injury significantly enhanced functional nerve recovery. Remarkably, following bilateral nerve transection, visible and functional nerve regeneration was observed in 7 out of 8 animals treated with FL2-siRNA, while no control-treated animals exhibited regeneration. These studies identify FL2 as a promising therapeutic target for enhancing regeneration after peripheral nerve injury and for mitigating neurogenic ED after RP - a condition for which, at present, only poor treatment options exist.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - David J. Sharp
- Department of Physiology and Biophysics
- Dominick P. Purpura Department of Neuroscience, and
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
29
|
Zhang X, Hashimoto JG, Han X, Zhang F, Linhardt RJ, Guizzetti M. Characterization of Glycosaminoglycan Disaccharide Composition in Astrocyte Primary Cultures and the Cortex of Neonatal Rats. Neurochem Res 2021; 46:595-610. [PMID: 33398638 PMCID: PMC9116028 DOI: 10.1007/s11064-020-03195-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/12/2020] [Accepted: 12/04/2020] [Indexed: 12/23/2022]
Abstract
Astrocytes are major producers of the extracellular matrix (ECM), which is involved in the plasticity of the developing brain. In utero alcohol exposure alters neuronal plasticity. Glycosaminoglycans (GAGs) are a family of polysaccharides present in the extracellular space; chondroitin sulfate (CS)- and heparan sulfate (HS)-GAGs are covalently bound to core proteins to form proteoglycans (PGs). Hyaluronic acid (HA)-GAGs are not bound to core proteins. In this study we investigated the contribution of astrocytes to CS-, HS-, and HA-GAG production by comparing the makeup of these GAGs in cortical astrocyte cultures and the neonatal rat cortex. We also explored alterations induced by ethanol in GAG and core protein levels in astrocytes. Finally, we investigated the relative expression in astrocytes of CS-PGs of the lectican family of proteins, major components of the brain ECM, in vivo using translating ribosome affinity purification (TRAP) (in Aldh1l1-EGFP-Rpl10a mice. Cortical astrocytes produce low levels of HA and show low expression of genes involved in HA biosynthesis compared to the whole developing cortex. Astrocytes have high levels of chondroitin-0-sulfate (C0S)-GAGs (possibly because of a higher sulfatase enzyme expression) and HS-GAGs. Ethanol upregulates C4S-GAGs as well as brain-specific lecticans neurocan and brevican, which are highly enriched in astrocytes of the developing cortex in vivo. These results begin to elucidate the role of astrocytes in the biosynthesis of CS- HS- and HA-GAGs, and suggest that ethanol-induced alterations of neuronal development may be in part mediated by increased astrocyte GAG levels and neurocan and brevican expression.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.
- VA Portland Health Care System, R&D39, 3710 SW Veterans Hospital Road, Portland, OR, 97239, USA.
| | - Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
- VA Portland Health Care System, R&D39, 3710 SW Veterans Hospital Road, Portland, OR, 97239, USA
| | - Xiaorui Han
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Fuming Zhang
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Robert J Linhardt
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.
- VA Portland Health Care System, R&D39, 3710 SW Veterans Hospital Road, Portland, OR, 97239, USA.
| |
Collapse
|
30
|
Nagase H, Higashi SL, Iweka CA, Pearson CS, Hirata Y, Geller HM, Katagiri Y. Reliable and sensitive detection of glycosaminoglycan chains with immunoblots. Glycobiology 2021; 31:116-125. [PMID: 32614944 PMCID: PMC7874388 DOI: 10.1093/glycob/cwaa060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 11/14/2022] Open
Abstract
Complex glycans play vital roles in many biological processes, ranging from intracellular signaling and organ development to tumor growth. Glycan expression is routinely assessed by the application of glycan-specific antibodies to cells and tissues. However, glycan-specific antibodies quite often show a large number of bands on immunoblots and it is hard to interpret the data when reliable controls are lacking. This limits the scope of glycobiology studies and poses challenges for replication. We sought to resolve this issue by developing a novel strategy that utilizes an immunoreaction enhancing technology to vastly improve the speed and quality of glycan-based immunoblots. As a representative case study, we used chondroitin sulfate glycosaminoglycan (CS-GAG) chains as the carbohydrate target and a monoclonal antibody, CS-56, as the probe. We discovered that preincubation of the antibody with its antigenic CS-GAG chain distinguishes true-positive signals from false-positive ones. We successfully applied this strategy to 10E4, a monoclonal anti heparan sulfate GAGs (HS-GAGs) antibody, where true-positive signals were confirmed by chemical HS-GAG depolymerization on the membrane. This evidence that glycan-specific antibodies can generate clear and convincing data on immunoblot with highly replicable results opens new opportunities for many facets of life science research in glycobiology.
Collapse
Affiliation(s)
- Haruna Nagase
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Sayuri L Higashi
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Chinyere A Iweka
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Craig S Pearson
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Yoko Hirata
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Yasuhiro Katagiri
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| |
Collapse
|
31
|
Jin W, Zhang F, Linhardt RJ. Glycosaminoglycans in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1325:189-204. [PMID: 34495536 DOI: 10.1007/978-3-030-70115-4_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glycosaminoglycans (GAGs) are linear polysaccharides that consist of alternating disaccharides sequences of uronic acids and/or galactose hexamino sugars most of which are sulfated. GAGs are ubiquitously expressed on the cell surface, in the intracellular milieu and in the extracellular matrix of all animal cells. Thus, GAGs exhibit many essential roles in a variety of physiological and pathological processes. The targets of GAGs are GAG-binding proteins and related proteins that are of significant interest to both the academic community and in the pharmaceutical industry. In this review, the structures of GAGs, their binding proteins, and analogs are presented that further the development of GAGs and their analogs for the treatment of neurodegenerative diseases agents.
Collapse
Affiliation(s)
- Weihua Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China.,Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA. .,Department of Biological Science, Departments of Chemistry and Chemical Biology and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.
| |
Collapse
|
32
|
Joffrin AM, Hsieh-Wilson LC. Photoaffinity Probes for the Identification of Sequence-Specific Glycosaminoglycan-Binding Proteins. J Am Chem Soc 2020; 142:13672-13676. [PMID: 32786811 DOI: 10.1021/jacs.0c06046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glycosaminoglycan (GAG)-protein interactions mediate critical physiological and pathological processes, such as neuronal plasticity, development, and viral invasion. However, mapping GAG-protein interaction networks is challenging as these interactions often require specific GAG sulfation patterns and involve transmembrane receptors or extracellular matrix-associated proteins. Here, we report the first GAG polysaccharide-based photoaffinity probes for the system-wide identification of GAG-binding proteins in living cells. A general platform for the modular, efficient assembly of various chondroitin sulfate (CS)-based photoaffinity probes was developed. Systematic evaluations led to benzophenone-containing probes that efficiently and selectively captured known CS-E-binding proteins in vitro and in cells. Importantly, the probes also enabled the identification of >50 new proteins from living neurons that interact with the neuroplasticity-relevant CS-E sulfation motif. Several candidates were independently validated and included membrane receptors important for axon guidance, innate immunity, synapse development, and synaptic plasticity. Overall, our studies provide a powerful approach for mapping GAG-protein interaction networks, revealing new potential functions for these polysaccharides and linking them to diseases such as Alzheimer's and autism.
Collapse
Affiliation(s)
- Amélie M Joffrin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, California 91125, United States
| | - Linda C Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, California 91125, United States
| |
Collapse
|
33
|
Khan SA, Mason RW, Kobayashi H, Yamaguchi S, Tomatsu S. Advances in glycosaminoglycan detection. Mol Genet Metab 2020; 130:101-109. [PMID: 32247585 PMCID: PMC7198342 DOI: 10.1016/j.ymgme.2020.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Glycosaminoglycans (GAGs) are negatively charged long linear (highly sulfated) polysaccharides consisting of repeating disaccharide units that are expressed on the surfaces of all nucleated cells. The expression of GAGs is required for embryogenesis, regulation of cell growth and proliferation, maintenance of tissue hydration, and interactions of the cells via receptors. Mucopolysaccharidoses (MPS) are caused by deficiency of specific lysosomal enzymes that result in the accumulation of GAGs in multiple tissues leading to organ dysfunction. Therefore, GAGs are important biomarkers for MPS. Without any treatment, patients with severe forms of MPS die within the first two decades of life. SCOPE OF REVIEW Accurate measurement of GAGs is important to understand the diagnosis and pathogenesis of MPS and to monitor therapeutic efficacy before, during, and after treatment of the disease. This review covers various qualitative and quantitative methods for measurement of GAGs, including dye specific, thin layer chromatography (TLC), capillary electrophoresis, high-performance liquid chromatography (HPLC), liquid chromatography-tandem mass spectrometry (LC-MS/MS), gas chromatography, ELISA, and automated high-throughput mass spectrometry. Major conclusion: There are several methods for GAG detection however, specific GAG detection in the various biological systems requires rapid, sensitive, specific, and cost-effective methods such as LC-MS/MS. GENERAL SIGNIFICANCE This review will describe different methods for GAG detection and analysis, including their advantages and limitation.
Collapse
Affiliation(s)
- Shaukat A Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | | | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University, Shimane, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Pediatrics, Shimane University, Shimane, Japan; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Alginate Sulfate Substrates Control Growth Factor Binding and Growth of Primary Neurons: Toward Engineered 3D Neural Networks. ACTA ACUST UNITED AC 2020; 4:e2000047. [DOI: 10.1002/adbi.202000047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/29/2020] [Indexed: 12/27/2022]
|
35
|
Li J, Wu C, Chu PK, Gelinsky M. 3D printing of hydrogels: Rational design strategies and emerging biomedical applications. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2020; 140:100543. [DOI: 10.1016/j.mser.2020.100543] [Citation(s) in RCA: 370] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
36
|
Reginensi D, Ortiz D, Pravia A, Burillo A, Morales F, Morgan C, Jimenez L, Dave KR, Perez-Pinzon MA, Gittens RA. Role of Region-Specific Brain Decellularized Extracellular Matrix on In Vitro Neuronal Maturation. Tissue Eng Part A 2020; 26:964-978. [PMID: 32103711 DOI: 10.1089/ten.tea.2019.0277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent advancements in tissue engineering suggest that biomaterials, such as decellularized extracellular matrix (ECM), could serve to potentiate the localization and efficacy of regenerative therapies in the central nervous system. Still, what factors and which mechanisms are required from these ECM-based biomaterials to exert their effect are not entirely understood. In this study, we use the brain as a novel model to test the effects of particular biochemical and structural properties by evaluating, for the first time, three different sections of the brain (i.e., cortex, cerebellum, and remaining areas) side-by-side and their corresponding decellularized counterparts using mechanical (4-day) and chemical (1-day) decellularization protocols. The three different brain subregions had considerably different initial conditions in terms of cell number and growth factor content, and some of these differences were maintained after decellularization. Decellularized ECM from both protocols was used as a substrate or as soluble factor, in both cases showing good cell attachment and growth capabilities. Interestingly, the 1-day protocol was capable of promoting greater differentiation than the 4-day protocol, probably due to its capacity to remove a similar amount of cell nuclei, while better conserving the biochemical and structural components of the cerebral ECM. Still, some limitations of this study include the need to evaluate the response in other biologically relevant cell types, as well as a more detailed characterization of the components in the decellularized ECM of the different brain subregions. In conclusion, our results show differences in neuronal maturation depending on the region of the brain used to produce the scaffolds. Complex organs such as the brain have subregions with very different initial cellular and biochemical conditions that should be considered for decellularization to minimize exposure to immunogenic components, while retaining bioactive factors conducive to regeneration. [Figure: see text] Impact statement The present study offers new knowledge about the production of decellularized extracellular matrix scaffolds from specific regions of the porcine brain, with a direct comparison of their effect on in vitro neuronal maturation. Our results show differences in neuronal maturation depending on the region of the brain used to produce the scaffolds, suggesting that it is necessary to consider the initial cellular content of the source tissue and its bioactive capacity for the production of an effective regenerative therapy for stroke.
Collapse
Affiliation(s)
- Diego Reginensi
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama.,School of Medicine, Universidad de Panamá, Panama, Republic of Panama.,Biomedical Engineering Program, Universidad Latina de Panamá, Panama, Republic of Panama
| | - Didio Ortiz
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Andrea Pravia
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama.,Biotechnology Program, Universidad Latina de Panamá, Panama, Republic of Panama
| | - Andrea Burillo
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Félix Morales
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Carly Morgan
- CREO-MIHRT Program, University of California, Santa Cruz, California.,Materials Science & Engineering Program, University of Washington, Seattle, Washington
| | - Lindsay Jimenez
- CREO-MIHRT Program, University of California, Santa Cruz, California.,Biomedical Engineering Program, University of Connecticut, Storrs, Connecticut
| | - Kunjan R Dave
- The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Neurology Department, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Miguel A Perez-Pinzon
- The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Neurology Department, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Rolando A Gittens
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama.,Centro de Biodiversidad y Descubrimiento de Drogas, INDICASAT AIP, City of Knowledge, Panama, Republic of Panama
| |
Collapse
|
37
|
Alhamoudi KM, Bhat J, Nashabat M, Alharbi M, Alyafee Y, Asiri A, Umair M, Alfadhel M. A Missense Mutation in the UGDH Gene Is Associated With Developmental Delay and Axial Hypotonia. Front Pediatr 2020; 8:71. [PMID: 32175296 PMCID: PMC7056728 DOI: 10.3389/fped.2020.00071] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/13/2020] [Indexed: 11/13/2022] Open
Abstract
UDP-glucose dehydrogenase (UGDH) encodes an oxidoreductase that converts two successive oxidations of UDP-glucose to produce UDP-glucuronic acid, a key component in the synthesis of several polysaccharides such as glycosaminoglycan and the disaccharide hyaluronic acid. UGDH is critical to the production of extracellular matrix components which are essential to the migration and connectivity of neurons early in human brain development. In this report, we describe one child of a consanguineous family who presented with distinct clinical features including global developmental delay, axial hypotonia, bilateral undescended testis, and subtle dysmorphic features. Whole genome sequencing and a segregation was performed to identify the genetic cause of the disease within the family. Though mutations in the UGDH protein have been described as causing developmental delay in various model organisms, to our knowledge, this is the first identification of the novel homozygous missense variant in exon8 of UGDH NM_003359.3: c.950 G>A (p.Arg317Gln) and most likely the cause of the patient's phenotype. This variant falls in an active region and replaces the highly conserved Arginine 317 residues across mammals.
Collapse
Affiliation(s)
- Kheloud M Alhamoudi
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Javaid Bhat
- Medical Core Facility and Research Platforms, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Marwan Nashabat
- Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Masheal Alharbi
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Yusra Alyafee
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulaziz Asiri
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Majid Alfadhel
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
38
|
Sun J, Zhang J, Li K, Zheng Q, Song J, Liang Z, Ding T, Qiao L, Zhang J, Hu X, Wang Z. Photobiomodulation Therapy Inhibit the Activation and Secretory of Astrocytes by Altering Macrophage Polarization. Cell Mol Neurobiol 2020; 40:141-152. [PMID: 31446561 PMCID: PMC11448964 DOI: 10.1007/s10571-019-00728-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) stimulates reactive astrogliosis and the infiltration of macrophages, which interact with each other at the injured area. We previously found Photobiomodulation (PBM) significantly decreases the number of M1 macrophages at the injured area of SCI. But the exact nature of the astrocyte response following PBM and relationship with the macrophage have not been explored in detail. In this study, a BALB/c mice model with standardized bilateral spinal cord compression and a macrophage-astrocyte co-culture model were applied to study effects of PBM on astrocytes. Results showed that PBM inhibit the expression of the astrocyte markers glial fibrillary acidic protein (GFAP) and the secretion of chondroitin sulfate proteoglycans (CSPG) in the para-epicenter area, decrease the number of M1 macrophage in vivo. The in vitro experiments indicated M1 macrophages promote the cell viability of astrocytes and the expression of CSPG. However, PBM significantly inhibited the expression of GFAP, decreased activation of astrocyte, and downregulated the expression of CSPG by regulating M1 macrophages. These results demonstrate that PBM may regulate the interaction between macrophages and astrocytes after spinal cord injury, which inhibited the formation of glial scar.
Collapse
Affiliation(s)
- Jiakai Sun
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Jiawei Zhang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Kun Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Qiao Zheng
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Jiwei Song
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Zhuowen Liang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Tan Ding
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China
| | - Lin Qiao
- Department of Orthopedics, Third Hospital of Chinese PLA, Baoji, Shaanxi, China
| | - Jianxin Zhang
- Department of Orthopedics, Weinan Central Hospital, Weinan, Shaanxi, China
| | - Xueyu Hu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China.
| | - Zhe Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
39
|
Fletcher EJR, Moon LDF, Duty S. Chondroitinase ABC reduces dopaminergic nigral cell death and striatal terminal loss in a 6-hydroxydopamine partial lesion mouse model of Parkinson's disease. BMC Neurosci 2019; 20:61. [PMID: 31862005 PMCID: PMC6923832 DOI: 10.1186/s12868-019-0543-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/10/2019] [Indexed: 11/28/2022] Open
Abstract
Background Parkinson’s disease (PD) is characterised by dopaminergic cell loss within the substantia nigra pars compacta (SNc) that leads to reduced striatal dopamine content and resulting motor deficits. Identifying new strategies to protect these cells from degeneration and retain striatal dopaminergic innervation is therefore of great importance. Chondroitin sulphate proteoglycans (CSPGs) are recognised contributors to the inhibitory extracellular milieu known to hinder tissue recovery following CNS damage. Digestion of these molecules by the bacterial lyase chondroitinase ABC (ChABC) has been shown to promote functional recovery in animal models of neurological injury. Although ChABC has been shown to promote sprouting of dopaminergic axons following transection of the nigrostriatal pathway, its ability to protect against nigrostriatal degeneration in a toxin-based module with better construct validity for PD has yet to be explored. Here we examined the neuroprotective efficacy of ChABC treatment in the full and partial 6-hydroxydopamine (6-OHDA) lesion mouse models of PD. Results In mice bearing a full 6-OHDA lesion, ChABC treatment failed to protect against the loss of either nigral cells or striatal terminals. In contrast, in mice bearing a partial 6-OHDA lesion, ChABC treatment significantly protected cells of the rostral SNc, which remained at more than double the numbers seen in vehicle-treated animals. In the partial lesion model, ChABC treatment also significantly preserved dopaminergic fibres of the rostral dorsal striatum which increased from 15.3 ± 3.5% of the intact hemisphere in saline-treated animals to 36.3 ± 6.5% in the ChABC-treated group. These protective effects of ChABC treatment were not accompanied by improvements in either the cylinder or amphetamine-induced rotations tests of motor function. Conclusions ChABC treatment provided significant protection against a partial 6-OHDA lesion of the nigrostriatal tract although the degree of protection was not sufficient to improve motor outcomes. These results support further investigations into the benefits of ChABC treatment for providing neuroprotection in PD.
Collapse
Affiliation(s)
- Edward J R Fletcher
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK
| | - Lawrence D F Moon
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK
| | - Susan Duty
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
40
|
Raghunathan R, Sethi MK, Klein JA, Zaia J. Proteomics, Glycomics, and Glycoproteomics of Matrisome Molecules. Mol Cell Proteomics 2019; 18:2138-2148. [PMID: 31471497 PMCID: PMC6823855 DOI: 10.1074/mcp.r119.001543] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
The most straightforward applications of proteomics database searching involve intracellular proteins. Although intracellular gene products number in the thousands, their well-defined post-translational modifications (PTMs) makes database searching practical. By contrast, cell surface and extracellular matrisome proteins pass through the secretory pathway where many become glycosylated, modulating their physicochemical properties, adhesive interactions, and diversifying their functions. Although matrisome proteins number only a few hundred, their high degree of complex glycosylation multiplies the number of theoretical proteoforms by orders of magnitude. Given that extracellular networks that mediate cell-cell and cell-pathogen interactions in physiology depend on glycosylation, it is important to characterize the proteomes, glycomes, and glycoproteomes of matrisome molecules that exist in a given biological context. In this review, we summarize proteomics approaches for characterizing matrisome molecules, with an emphasis on applications to brain diseases. We demonstrate the availability of methods that should greatly increase the availability of information on matrisome molecular structure associated with health and disease.
Collapse
Affiliation(s)
- Rekha Raghunathan
- Molecular and Translational Medicine Program, Boston University, Boston, MA 02218; Department of Biochemistry, Boston University, Boston, MA 02218
| | - Manveen K Sethi
- Department of Biochemistry, Boston University, Boston, MA 02218
| | - Joshua A Klein
- Bioinformatics Program, Boston University, Boston, MA 02218
| | - Joseph Zaia
- Molecular and Translational Medicine Program, Boston University, Boston, MA 02218; Department of Biochemistry, Boston University, Boston, MA 02218; Bioinformatics Program, Boston University, Boston, MA 02218.
| |
Collapse
|
41
|
Neuronal Pentraxin 2 Binds PNNs and Enhances PNN Formation. Neural Plast 2019; 2019:6804575. [PMID: 31772567 PMCID: PMC6854953 DOI: 10.1155/2019/6804575] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023] Open
Abstract
The perineuronal net (PNN) is a mesh-like proteoglycan structure on the neuronal surface which is involved in regulating plasticity. The PNN regulates plasticity via multiple pathways, one of which is direct regulation of synapses through the control of AMPA receptor mobility. Since neuronal pentraxin 2 (Nptx2) is a known regulator of AMPA receptor mobility and Nptx2 can be removed from the neuronal surface by PNN removal, we investigated whether Nptx2 has a function in the PNN. We found that Nptx2 binds to the glycosaminoglycans hyaluronan and chondroitin sulphate E in the PNN. Furthermore, in primary cortical neuron cultures, the addition of NPTX2 to the culture medium enhances PNN formation during PNN development. These findings suggest Nptx2 as a novel PNN binding protein with a role in the mechanism of PNN formation.
Collapse
|
42
|
Malaeb W, Bahmad HF, Abou-Kheir W, Mhanna R. The sulfation of biomimetic glycosaminoglycan substrates controls binding of growth factors and subsequent neural and glial cell growth. Biomater Sci 2019; 7:4283-4298. [PMID: 31407727 DOI: 10.1039/c9bm00964g] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sulfated glycosaminoglycans (GAGs) are key structural and functional extracellular matrix (ECM) molecules involved in numerous signaling pathways mainly through their interaction with growth factors. Alginate sulfate mimics sulfated GAGs and binds growth factors such as basic fibroblast growth factor (FGF-2). Here, natural biomimetic substrates were engineered by immobilizing biotinylated alginate sulfates with varying degrees of sulfation (DS, from 0 to 2.7) on gold and polystyrene substrates using biotin-streptavidin binding. The build-up of films and the effect of the DS and biotinylation method on FGF-2 binding were assessed using quartz crystal microbalance with dissipation monitoring (QCM-D) and immunohistochemistry. The role of substrate sulfation and FGF-2 loading on the growth of A172 (human glioblastoma multiforme), SH-SY5Y (human neuroblastoma), and PC-12 (rat pheochromocytoma) cell lines was evaluated in vitro using proliferation and neurite outgrowth assessment. An increase in the DS of alginates resulted in augmented FGF-2 binding as evidenced by higher frequency and dissipation shifts measured with QCM-D and confirmed with immunostaining. All sulfated alginate substrates supported the attachment and growth of neural/glial cell lines better than controls with the highest increase in cell proliferation observed for the highest DS (p < 0.05 for all the cell lines). Moreover, FGF-2 loaded substrates with the highest DS induced the most significant increase in neurite-positive PC-12 cells and average neurite length. The developed biomimetic coatings can be used to functionalize substrates for biosensing applications (e.g. gold substrates) and to induce defined cellular responses via controlled growth factor delivery for basic and applied sciences.
Collapse
Affiliation(s)
- Waddah Malaeb
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Hisham F Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| |
Collapse
|
43
|
Krishnaswamy VR, Benbenishty A, Blinder P, Sagi I. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell Mol Life Sci 2019; 76:3229-3248. [PMID: 31197404 PMCID: PMC11105229 DOI: 10.1007/s00018-019-03182-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022]
Abstract
The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.
Collapse
Affiliation(s)
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Blinder
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
44
|
Pomin VH, Vignovich WP, Gonzales AV, Vasconcelos AA, Mulloy B. Galactosaminoglycans: Medical Applications and Drawbacks. Molecules 2019; 24:E2803. [PMID: 31374852 PMCID: PMC6696379 DOI: 10.3390/molecules24152803] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 12/28/2022] Open
Abstract
Galactosaminoglycans (GalAGs) are sulfated glycans composed of alternating N-acetylgalactosamine and uronic acid units. Uronic acid epimerization, sulfation patterns and fucosylation are modifications observed on these molecules. GalAGs have been extensively studied and exploited because of their multiple biomedical functions. Chondroitin sulfates (CSs), the main representative family of GalAGs, have been used in alternative therapy of joint pain/inflammation and osteoarthritis. The relatively novel fucosylated chondroitin sulfate (FCS), commonly found in sea cucumbers, has been screened in multiple systems in addition to its widely studied anticoagulant action. Biomedical properties of GalAGs are directly dependent on the sugar composition, presence or lack of fucose branches, as well as sulfation patterns. Although research interest in GalAGs has increased considerably over the three last decades, perhaps motivated by the parallel progress of glycomics, serious questions concerning the effectiveness and potential side effects of GalAGs have recently been raised. Doubts have centered particularly on the beneficial functions of CS-based therapeutic supplements and the potential harmful effects of FCS as similarly observed for oversulfated chondroitin sulfate, as a contaminant of heparin. Unexpected components were also detected in CS-based pharmaceutical preparations. This review therefore aims to offer a discussion on (1) the current and potential therapeutic applications of GalAGs, including those of unique features extracted from marine sources, and (2) the potential drawbacks of this class of molecules when applied to medicine.
Collapse
Affiliation(s)
- Vitor H Pomin
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA.
- Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA.
| | - William P Vignovich
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA
| | - Alysia V Gonzales
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA
| | - Ariana A Vasconcelos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Barbara Mulloy
- Imperial College, Department of Medicine, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
45
|
Liu LC, Wang YL, Lin PL, Zhang X, Cheng WC, Liu SH, Chen CJ, Hung Y, Jan CI, Chang LC, Qi X, Hsieh-Wilson LC, Wang SC. Long noncoding RNA HOTAIR promotes invasion of breast cancer cells through chondroitin sulfotransferase CHST15. Int J Cancer 2019; 145:2478-2487. [PMID: 30963568 DOI: 10.1002/ijc.32319] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/04/2019] [Accepted: 03/25/2019] [Indexed: 12/28/2022]
Abstract
The long noncoding RNA HOTAIR plays significant roles in promoting cancer metastasis. However, how it conveys an invasive advantage in cancer cells is not clear. Here we identify the chondroitin sulfotransferase CHST15 (GalNAc4S-6ST) as a novel HOX transcript antisense intergenic RNA (HOTAIR) target gene using RNA profiling and show that CHST15 is required for HOTAIR-mediated invasiveness in breast cancer cells. CHST15 catalyzes sulfation of the C6 hydroxyl group of the N-acetyl galactosamine 4-sulfate moiety in chondroitin sulfate to form the 4,6-disulfated chondroitin sulfate variant known as the CS-E isoform. We show that HOTAIR is necessary and sufficient for CHST15 transcript expression. Inhibition of CHST15 by RNA interference abolished cell invasion promoted by HOTAIR but not on HOTAIR-mediated migratory activity. Conversely, reconstitution of CHST15 expression rescued the invasive activity of HOTAIR-depleted cells. In corroboration with this mechanism, blocking cell surface chondroitin sulfate using a pan-CS antibody or an antibody specifically recognizes the CS-E isoform significantly suppressed HOTAIR-induced invasion. Inhibition of CHST15 compromised tumorigenesis and metastasis in orthotopic breast cancer xenograft models. Furthermore, the expression of HOTAIR closely correlated with the level of CHST15 protein in primary as well as metastatic tumor lesions. Our results demonstrate a novel mechanism underlying the function of HOTAIR in tumor progression through programming the context of cell surface glycosaminoglycans. Our results further establish that the invasive and migratory activities downstream of HOTAIR are distinctly regulated, whereby CHST15 preferentially controls the arm of invasiveness. Thus, the HOTAIR-CHST15 axis may provide a new avenue toward novel therapeutic strategies and prognosis biomarkers for advanced breast cancer.
Collapse
Affiliation(s)
- Liang-Chih Liu
- Department of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Liang Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Le Lin
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati, Cincinnati, OH
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Shu-Hsuan Liu
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Jung Chen
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yu Hung
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Ing Jan
- Division of Molecular Pathology, Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Ling-Chu Chang
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Xiaoyang Qi
- Department of Hematology Oncology, University of Cincinnati, Cincinnati, OH
| | - Linda C Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of Cancer Biology, University of Cincinnati, Cincinnati, OH.,Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
46
|
Loers G, Liao Y, Hu C, Xue W, Shen H, Zhao W, Schachner M. Identification and characterization of synthetic chondroitin-4-sulfate binding peptides in neuronal functions. Sci Rep 2019; 9:1064. [PMID: 30705359 PMCID: PMC6355858 DOI: 10.1038/s41598-018-37685-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/28/2018] [Indexed: 02/05/2023] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs), up-regulated in and around the glial scar after mammalian spinal cord injury, have been suggested to be key inhibitory molecules for functional recovery by impeding axonal regrowth/sprouting and synaptic rearrangements. CSPG-mediated inhibition is mainly associated with the glycosaminoglycan chains of CSPGs, and chondroitin-4-sulfate (C4S) is the predominant sulfated structure that regulates axonal guidance and growth in the adult nervous system. With the aim to find molecules that neutralize the inhibitory functions of C4S, we screened a phage display library for peptides binding to C4S. From the phage clones binding to C4S we selected three peptides for further analysis. We observed that these peptides bind to C4S, but not chondroitin-6-sulfate, heparin sulfate or dermatan sulfate, in a concentration-dependent and saturable manner, whereas the scrambled peptides showed highly reduced or no binding to C4S. The C4S-binding peptides, but not their scrambled counterparts, when added to cultures of mouse cerebellar neurons and human neuroblastoma cells, neutralized the inhibitory functions of the C4S- and CSPG-coated substrate on cell adhesion, neuronal migration and neurite outgrowth. These results indicate that the C4S-binding peptides neutralize several inhibitory functions of CSPGs, suggesting that they may be beneficial in repairing mammalian nervous system injuries.
Collapse
Affiliation(s)
- Gabriele Loers
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Yonghong Liao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Chengliang Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Weikang Xue
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Huifan Shen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Weijiang Zhao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China.
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China.
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
47
|
Makyła-Juzak K, Chachaj-Brekiesz A, Dynarowicz-Latka P, Dąbczyński P, Zemla J. The Effect of Dextran Sulfate-as Model Glycosaminoglycan Analogue-on Membrane Lipids: DPPC, Cholesterol, and DPPC-Cholesterol Mixture. The Monolayer Study. J Membr Biol 2018; 251:641-651. [PMID: 30030544 PMCID: PMC6244761 DOI: 10.1007/s00232-018-0041-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
Abstract
Glycosaminoglycans (GAGs) are essential components of the extracellular matrices (ECMs) located on the outer surface of cellular membranes. They belong to the group of polysaccharides involved in diverse biological processes acting on the surface and across natural lipid membranes. Recently, particular attention has been focused on possible role of GAGs in the amyloid deposits. The amyloid formation is related to a disorder in protein folding, causing that soluble-in normal conditions-peptides become deposited extracellularly as insoluble fibrils, impairing tissue structure and its function. One of the hypothesis holds that GAGs may inhibit amyloid formation by interacting with the lipid membrane by blocking the accumulation of protein aggregates on the membrane surface. Although the biophysical properties of GAGs are described rather well, little is known about the nature of association between these polysaccharides and components of natural cell membranes. Therefore, a study of GAGs influence on membrane lipids is of particular importance. The aim of the present work is to get insight into the effect of hydrophilic dextran sulfate (DS)-that can be considered as GAG analogue-on membrane lipids organization. This study was based on examining interactions between DS sodium salt of molecular weight equal to about 40 kDa (DS40), dissolved in water subphase, and a model membrane, mimicked as Langmuir monolayer, formed by representative natural membrane lipids: cholesterol and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) as well as their mixtures. Due to the fact that calcium ions in excess may accumulate in the lipid membrane, attracting high molecular weight molecules to their surface, the influence of calcium ions present in the subphase on the DS40 activity has also been examined. It has been found that negatively charged DS, forming a sublayer underneath the monolayer, barely interacts with membrane lipids; however, in the presence of calcium ions the electrostatic interactions between DS40 and lipid membrane are significantly enhanced, leading to the formation of network-like crystalline structures at the surface of model membrane, which can prevent incorporation and interaction with other extracellular molecules, e.g., proteins.
Collapse
Affiliation(s)
- Katarzyna Makyła-Juzak
- Department of General Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland.
| | - Anna Chachaj-Brekiesz
- Department of General Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| | - Patrycja Dynarowicz-Latka
- Department of General Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| | - Paweł Dąbczyński
- Institute of Physics, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Joanna Zemla
- Institute of Nuclear Physics, Polish Academy of Sciences, Radzikowskiego 152, 31-342, Kraków, Poland
| |
Collapse
|
48
|
Jiang B, Xu F, Li L, Chen W, Hong S, Chen R. The inhibition of glycosaminoglycan incorporation influences the cell proliferation and cytodifferentiation in cultured embryonic mouse molars. J Mol Histol 2018; 50:11-19. [PMID: 30498999 DOI: 10.1007/s10735-018-9803-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 11/08/2018] [Indexed: 01/19/2023]
Abstract
The extracellular matrix (ECM) contains a variety of complex macromolecules including proteoglycans (PGs) and glycosaminoglycans (GAGs). PG consists of a protein core with covalently attached carbohydrate side chains called GAGs. Several PGs, including versican, biglycan, decorin and syndecan are involved in odontogenesis while the role of GAGs in those PGs in this process remains unclarified. The purpose of this study was to investigate the influence of GAGs on tooth development. The mandibular first molars at early bell stage were cultivated with or without 4-methylumbelliferyl-β-D-xyloside (Xyl-MU). The cultured tooth germs were metabolically labelled with [35S] Na2SO4, then PGs in tooth germs and cultured medium were extracted separately and analyzed by gel filtration. Morphological changes were evaluated on days 2, 4, 6, and histological changes were examined by hematoxylin-eosin (HE) staining and transmission electron microscope (TEM). Related proteins and genes of cytodifferentiation were further examined by immunohistochemistry (IHC) and quantitive real-time PCR (qPCR) respectively. Meanwhile, BrdU incorporation assay was used to explore the effect of Xyl-MU on the cell proliferation of cultured tooth germs. The results demonstrated that the incorporation of GAGs to PGs in cultured tooth germs was heavily inhibited by Xyl-MU. Accompanied by the inhibition of GAGs incorporation, Xyl-MU altered tooth morphogenesis and delayed the differentiation of ameloblasts and odontoblasts. Proliferation of inner enamel epithelium (IEE) was also inhibited. Therefore, we draw a conclusion that the inhibition of GAGs incorporation influences the cell proliferation and cytodifferentiation in cultured embryonic mouse molars.
Collapse
Affiliation(s)
- Beizhan Jiang
- Department of Operative Dentistry and Endodontics, School & Hosipital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai, 200072, China.
| | - Fangfang Xu
- Department of Operative Dentistry and Endodontics, School & Hosipital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Lefeng Li
- Department of Operative Dentistry and Endodontics, School & Hosipital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Weiting Chen
- Department of Operative Dentistry and Endodontics, School & Hosipital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Shebin Hong
- Department of Operative Dentistry and Endodontics, School & Hosipital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Rongmei Chen
- Department of Operative Dentistry and Endodontics, School & Hosipital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 399 Middle Yan Chang Road, Shanghai, 200072, China
| |
Collapse
|
49
|
García-Bernal F, Geribaldi-Doldán N, Domínguez-García S, Carrasco M, Murillo-Carretero M, Delgado-Ariza A, Díez-Salguero M, Verástegui C, Castro C. Protein Kinase C Inhibition Mediates Neuroblast Enrichment in Mechanical Brain Injuries. Front Cell Neurosci 2018; 12:462. [PMID: 30542270 PMCID: PMC6277931 DOI: 10.3389/fncel.2018.00462] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/13/2018] [Indexed: 01/08/2023] Open
Abstract
Brain injuries of different etiologies lead to irreversible neuronal loss and persisting neuronal deficits. New therapeutic strategies are emerging to compensate neuronal damage upon brain injury. Some of these strategies focus on enhancing endogenous generation of neurons from neural stem cells (NSCs) to substitute the dying neurons. However, the capacity of the injured brain to produce new neurons is limited, especially in cases of extensive injury. This reduced neurogenesis is a consequence of the effect of signaling molecules released in response to inflammation, which act on intracellular pathways, favoring gliogenesis and preventing recruitment of neuroblasts from neurogenic regions. Protein kinase C (PKC) is a family of intracellular kinases involved in several of these gliogenic signaling pathways. The aim of this study was to analyze the role of PKC isozymes in the generation of neurons from neural progenitor cells (NPCs) in vitro and in vivo in brain injuries. PKC inhibition in vitro, in cultures of NPC isolated from the subventricular zone (SVZ) of postnatal mice, leads differentiation towards a neuronal fate. This effect is not mediated by classical or atypical PKC. On the contrary, this effect is mediated by novel PKCε, which is abundantly expressed in NPC cultures under differentiation conditions. PKCε inhibition by siRNA promotes neuronal differentiation and reduces glial cell differentiation. On the contrary, inhibition of PKCθ exerts a small anti-gliogenic effect and reverts the effect of PKCε inhibition on neuronal differentiation when both siRNAs are used in combination. Interestingly, in cortical brain injuries we have found expression of almost all PKC isozymes found in vitro. Inhibition of PKC activity in this type of injuries leads to neuronal production. In conclusion, these findings show an effect of PKCε in the generation of neurons from NPC in vitro, and they highlight the role of PKC isozymes as targets to produce neurons in brain lesions.
Collapse
Affiliation(s)
- Francisco García-Bernal
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Noelia Geribaldi-Doldán
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Manuel Carrasco
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Maribel Murillo-Carretero
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | | | - Mónica Díez-Salguero
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Cristina Verástegui
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Departamento de Anatomía, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| |
Collapse
|
50
|
Wellman SM, Cambi F, Kozai TD. The role of oligodendrocytes and their progenitors on neural interface technology: A novel perspective on tissue regeneration and repair. Biomaterials 2018; 183:200-217. [PMID: 30172245 PMCID: PMC6469877 DOI: 10.1016/j.biomaterials.2018.08.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
Oligodendrocytes and their precursors are critical glial facilitators of neurophysiology, which is responsible for cognition and behavior. Devices that are used to interface with the brain allow for a more in-depth analysis of how neurons and these glia synergistically modulate brain activity. As projected by the BRAIN Initiative, technologies that acquire a high resolution and robust sampling of neural signals can provide a greater insight in both the healthy and diseased brain and support novel discoveries previously unobtainable with the current state of the art. However, a complex series of inflammatory events triggered during device insertion impede the potential applications of implanted biosensors. Characterizing the biological mechanisms responsible for the degradation of intracortical device performance will guide novel biomaterial and tissue regenerative approaches to rehabilitate the brain following injury. Glial subtypes which assist with neuronal survival and exchange of electrical signals, mainly oligodendrocytes, their precursors, and the insulating myelin membranes they produce, are sensitive to inflammation commonly induced from insults to the brain. This review explores essential physiological roles facilitated by oligodendroglia and their precursors and provides insight into their pathology following neurodegenerative injury and disease. From this knowledge, inferences can be made about the impact of device implantation on these supportive glia in order to engineer effective strategies that can attenuate their responses, enhance the efficacy of neural interfacing technology, and provide a greater understanding of the challenges that impede wound healing and tissue regeneration during pathology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, USA
| | - Takashi Dy Kozai
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, USA; NeuroTech Center, University of Pittsburgh Brain Institute, USA.
| |
Collapse
|