1
|
John RK, Vogel SP, Zia S, Lee KV, Nguyen AT, Torres-Espin A, Fenrich KK, Ng C, Schmidt EKA, Vavrek R, Raposo PJF, Smith K, Fouad K, Plemel JR. Reawakening inflammation in the chronically injured spinal cord using lipopolysaccharide induces diverse microglial states. J Neuroinflammation 2025; 22:56. [PMID: 40022205 PMCID: PMC11871772 DOI: 10.1186/s12974-025-03379-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Rehabilitative training is an effective method to promote recovery following spinal cord injury (SCI), with lower training efficacy observed in the chronic stage. The increased training efficacy during the subacute period is associated with a shift towards a more adaptive or proreparative state induced by the SCI. A potential link is SCI-induced inflammation, which is elevated in the subacute period, and, as injection of lipopolysaccharide (LPS) alongside training improves recovery in chronic SCI, suggesting LPS could reopen a window of plasticity late after injury. Microglia may play a role in LPS-mediated plasticity as they react to LPS and are implicated in facilitating recovery following SCI. However, it is unknown how microglia change in response to LPS following SCI to promote neuroplasticity. MAIN BODY Here we used single-cell RNA sequencing to examine microglial responses in subacute and chronic SCI with and without an LPS injection. We show that subacute SCI is characterized by a disease-associated microglial (DAM) signature, while chronic SCI is highly heterogeneous, with both injury-induced and homeostatic states. DAM states exhibit predicted metabolic pathway activity and neuronal interactions that are associated with potential mediators of plasticity. With LPS injection, microglia shifted away from the homeostatic signature to a primed, translation-associated state and increased DAM in degenerated tracts caudal to the injury. CONCLUSION Microglial states following an inflammatory stimulus in chronic injury incompletely recapitulate the subacute injury environment, showing both overlapping and distinct microglial signatures across time and with LPS injection. Our results contribute to an understanding of how microglia and LPS-induced neuroinflammation contribute to plasticity following SCI.
Collapse
Affiliation(s)
- Rebecca K John
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sadie P Vogel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Kelly V Lee
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Antoinette T Nguyen
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Abel Torres-Espin
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Carmen Ng
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Emma K A Schmidt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Pamela J F Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Keira Smith
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada.
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| |
Collapse
|
2
|
Kheirollahi A, Sadeghi S, Orandi S, Moayedi K, Khajeh K, Khoobi M, Golestani A. Chondroitinase as a therapeutic enzyme: Prospects and challenges. Enzyme Microb Technol 2024; 172:110348. [PMID: 37898093 DOI: 10.1016/j.enzmictec.2023.110348] [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: 05/22/2023] [Revised: 09/28/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
The chondroitinases (Chase) are bacterial lyases that specifically digest chondroitin sulfate and/or dermatan sulfate glycosaminoglycans via a β-elimination reaction and generate unsaturated disaccharides. In recent decades, these enzymes have attracted the attention of many researchers due to their potential applications in various aspects of medicine from the treatment of spinal cord injury to use as an analytical tool. In spite of this diverse spectrum, the application of Chase is faced with several limitations and challenges such as thermal instability and lack of a suitable delivery system. In the current review, we address potential therapeutic applications of Chase with emphasis on the challenges ahead. Then, we summarize the latest achievements to overcome the problems by considering the studies carried out in the field of enzyme engineering, drug delivery, and combination-based therapy.
Collapse
Affiliation(s)
- Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Solmaz Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Orandi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiana Moayedi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Silva MB, Pinto LDLDS, Medeiros LH, Souza AA, Chavante SF, Filgueira LGA, Camara RBG, Sassaki GL, Rocha HAO, Andrade GPV. Chondroitin Sulfate from Oreochromis niloticus Waste Reduces Leukocyte Influx in an Acute Peritonitis Model. Molecules 2023; 28:molecules28073082. [PMID: 37049845 PMCID: PMC10096408 DOI: 10.3390/molecules28073082] [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: 02/23/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Oreochromis niloticus (tilapia) is one of the most cultivated fish species worldwide. Tilapia farming generates organic waste from fish removal processes in nurseries. Visceral waste can damage natural ecosystems. Therefore, the use of this material as a source of biomolecules helps reduce environmental impacts and improve pharmacological studies. Tilapia viscera were subjected to proteolysis and complexation with an ion-exchange resin. The obtained glycosaminoglycans were purified using ion exchange chromatography (DEAE-Sephacel). The electrophoretic profile and analysis of 1H/13C nuclear magnetic resonance (NMR) spectra allowed for the characterization of the compound as chondroitin sulfate and its sulfation position. This chondroitin was named CST. We tested the ability of CST to reduce leukocyte influx in acute peritonitis models induced by sodium thioglycolate and found a significant reduction in leukocyte migration to the peritoneal cavity, similar to the polymorphonuclear population of the three tested doses of CST. This study shows, for the first time, the potential of CST obtained from O. niloticus waste as an anti-inflammatory drug, thereby contributing to the expansion of the study of molecules with pharmacological functions.
Collapse
Affiliation(s)
- Marianna Barros Silva
- Laboratório de Biotecnologia de Polímeros Naturais (BIOPOL), Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Lívia de Lourdes de Sousa Pinto
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Luiz Henrique Medeiros
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Airton Araújo Souza
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Norte, Campus de Parnamirim, Parnamirim 59143-455, RN, Brazil
| | - Suely Ferreira Chavante
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Luciana Guimarães Alves Filgueira
- Laboratório de Biotecnologia de Polímeros Naturais (BIOPOL), Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Rafael Barros Gomes Camara
- Laboratório de Biotecnologia de Polímeros Naturais (BIOPOL), Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Guilherme Lanzi Sassaki
- Departamento de Bioquímica e Biologia Molecular, Setor de Ciências Biológicas, Universidade Federal do Parana (UFPR), Curitiba 81531-980, PR, Brazil
| | - Hugo Alexandre Oliveira Rocha
- Laboratório de Biotecnologia de Polímeros Naturais (BIOPOL), Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Giulianna Paiva Viana Andrade
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| |
Collapse
|
4
|
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]
|
5
|
Pintér P, Alpár A. The Role of Extracellular Matrix in Human Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms231911085. [PMID: 36232390 PMCID: PMC9569603 DOI: 10.3390/ijms231911085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The dense neuropil of the central nervous system leaves only limited space for extracellular substances free. The advent of immunohistochemistry, soon followed by advanced diagnostic tools, enabled us to explore the biochemical heterogeneity and compartmentalization of the brain extracellular matrix in exploratory and clinical research alike. The composition of the extracellular matrix is critical to shape neuronal function; changes in its assembly trigger or reflect brain/spinal cord malfunction. In this study, we focus on extracellular matrix changes in neurodegenerative disorders. We summarize its phenotypic appearance and biochemical characteristics, as well as the major enzymes which regulate and remodel matrix establishment in disease. The specifically built basement membrane of the central nervous system, perineuronal nets and perisynaptic axonal coats can protect neurons from toxic agents, and biochemical analysis revealed how the individual glycosaminoglycan and proteoglycan components interact with these molecules. Depending on the site, type and progress of the disease, select matrix components can either proactively trigger the formation of disease-specific harmful products, or reactively accumulate, likely to reduce tissue breakdown and neuronal loss. We review the diagnostic use and the increasing importance of medical screening of extracellular matrix components, especially enzymes, which informs us about disease status and, better yet, allows us to forecast illness.
Collapse
Affiliation(s)
- Panka Pintér
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, 1051 Budapest, Hungary
- Correspondence:
| |
Collapse
|
6
|
YsHyl8A, an Alkalophilic Cold-Adapted Glycosaminoglycan Lyase Cloned from Pathogenic Yersinia sp. 298. Molecules 2022; 27:molecules27092897. [PMID: 35566248 PMCID: PMC9105423 DOI: 10.3390/molecules27092897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
A high enzyme-yield strain Yersinia sp. 298 was screened from marine bacteria harvested from the coastal water. The screening conditions were extensive, utilizing hyaluronic acid (HA)/chondroitin sulfate (CS) as the carbon source. A coding gene yshyl8A of the family 8 polysaccharide lyase (PL8) was cloned from the genome of Yersinia sp. 298 and subjected to recombinant expression. The specific activity of the recombinase YsHyl8A was 11.19 U/mg, with an optimal reaction temperature of 40 °C and 50% of its specific activity remaining after thermal incubation at 30 °C for 1 h. In addition, its optimal reaction pH was 7.5, and while it was most stable at pH 6.0 in Na2HPO4-citric acid buffer, it remained highly stable at pH 6.0–11.0. Further, its enzymatic activity was increased five-fold with 0.1 M NaCl. YsHyl8A, as an endo-lyase, can degrade both HA and CS, producing disaccharide end-products. These properties suggested that YsHyl8A possessed both significant alkalophilic and cold-adapted features while being dependent on NaCl, likely resulting from its marine source. Yersinia is a typical fish pathogen, with glycosaminoglycan lyase (GAG lyase) as a potential pathogenic factor, exhibiting strong hyaluronidase and chondroitinase activity. Further research on the pathogenic mechanism of GAG lyase may benefit the prevention and treatment of related diseases.
Collapse
|
7
|
Park HH, Kim YM, Anh Hong LT, Kim HS, Hoon KS, Jin X, Hwang DH, Kwon MJ, Song SC, Kim BG. Dual-functional hydrogel system for spinal cord regeneration with sustained release of arylsulfatase B alleviates fibrotic microenvironment and promotes axonal regeneration. Biomaterials 2022; 284:121526. [DOI: 10.1016/j.biomaterials.2022.121526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 12/20/2022]
|
8
|
Kumar A, Biswas A, Bojja SL, Kolathur KK, Volety SM. Emerging therapeutic role of chondroitinase (ChABC) in neurological disorders and cancer. CURRENT DRUG THERAPY 2022. [DOI: 10.2174/1574885517666220331151619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Proteoglycans are essential biomacromolecules that participate in matrix structure and organization, cell proliferation and migration, and cell surface signal transduction. However, their roles in physiology, particularly in CNS remain incompletely deciphered. Numerous studies highlight the elevated levels of chondroitin sulphate proteoglycans (CSPGs) in various diseases like cancers and neurological disorders like spinal cord injury (SCI), traumatic brain damage, neurodegenerative diseases, and are mainly implicated to hinder tissue repair. In such a context, chondroitinase ABC (ChABC), a therapeutic enzyme has shown immense hope to treat these diseases in several preclinical studies, primarily attributed to the digestion of the side chains of the proteoglycan chondroitin sulphate (CS) molecule. Despite extensive research, the progress in evolving the concept of therapeutic targeting of proteoglycans is still in its infancy. This review thus provides fresh insights into the emerging therapeutic applications of ChABC in various diseases apart from SCI and the underlying mechanisms.
Collapse
Affiliation(s)
- Akshara Kumar
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Aishi Biswas
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sree Lalitha Bojja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Kiran Kumar Kolathur
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Subrahmanyam M Volety
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
9
|
Inhibition of Autophagy Flux Promotes Secretion of Chondroitin Sulfate Proteoglycans in Primary Rat Astrocytes. Mol Neurobiol 2021; 58:6077-6091. [PMID: 34449046 DOI: 10.1007/s12035-021-02533-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
Following spinal cord injury (SCI), reactive astrocytes in the glial scar produce high levels of chondroitin sulfate proteoglycans (CSPGs), which are known to inhibit axonal regeneration. Transforming growth factor beta (TGFβ) is a well-known factor that induces the production of CSPGs, and in this study, we report a novel mechanism underlying TGFβ's effects on CSPG secretion in primary rat astrocytes. We observed increased TGFβ-induced secretion of the CSPGs neurocan and brevican, and this occurred simultaneously with inhibition of autophagy flux. In addition, we show that neurocan and brevican levels are further increased when TGFβ is administered in the presence of an autophagy inhibitor, Bafilomycin-A1, while they are reduced when cells are treated with a concentration of rapamycin that is not sufficient to induce autophagy. These findings suggest that TGFβ mediates its effects on CSPG secretion through autophagy pathways. They also represent a potential new approach to reduce CSPG secretion in vivo by targeting autophagy pathways, which could improve axonal regeneration after SCI.
Collapse
|
10
|
Ursavas S, Darici H, Karaoz E. Olfactory ensheathing cells: Unique glial cells promising for treatments of spinal cord injury. J Neurosci Res 2021; 99:1579-1597. [PMID: 33605466 DOI: 10.1002/jnr.24817] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/08/2021] [Indexed: 12/26/2022]
Abstract
Spinal cord injury (SCI) is generally the consequence of physical damage, which may result in devastating consequences such as paraplegia or paralysis. Some certain candidates for SCI repair are olfactory ensheathing cells (OECs), which are unique glial cells located in the transition region of the peripheral nervous system and central nervous system and perform neuron regeneration in the olfactory system throughout life. Culture studies have clarified many properties of OECs, but their mechanisms of actions are not fully understood. Successful results achieved in animal models showcased that SCI treatment with OEC transplants is suitable for clinical trials. However, clinical trials are limited by difficulties like cell acquisition for autograft transplantation. Despite the improvements in both animal and clinical studies so far, there is still insufficient information about the mechanism of actions, adverse effects, proper application methods, effective subtypes, and sources of cells. This review summarizes pre-clinical and clinical literature focused on the cellular characterization of both OECs in vitro and post-transplantation. We highlight the roles and effects of OECs on (a) the injury-induced glial milieu, (b) neuronal growth/regeneration, and (c) functional recovery after injury. Due to the shown benefits of OECs with in vitro and animal studies and a limited number of clinical trials, where safety and effectivity were shown, it is necessary to conduct more studies on OECs to obtain effective and feasible treatment methods.
Collapse
Affiliation(s)
- Selin Ursavas
- Department of Histology and Embryology, Faculty of Medicine, Istinye University, Istanbul, Turkey
| | - Hakan Darici
- Department of Histology and Embryology, Faculty of Medicine, Istinye University, Istanbul, Turkey
| | - Erdal Karaoz
- Department of Histology and Embryology, Faculty of Medicine, Istinye University, Istanbul, Turkey.,Center for Stem Cell and Tissue Engineering Research & Practice, Istinye University, Istanbul, Turkey.,Center for Regenerative Medicine and Stem Cell Research and Manufacturing, Liv Hospital, Istanbul, Turkey
| |
Collapse
|
11
|
Bertocchi I, Mele P, Ferrero G, Oberto A, Carulli D, Eva C. NPY-Y1 receptor signaling controls spatial learning and perineuronal net expression. Neuropharmacology 2020; 184:108425. [PMID: 33285203 DOI: 10.1016/j.neuropharm.2020.108425] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Perineuronal nets (PNNs) are extracellular matrix structures that form around some types of neurons at the end of critical periods, limiting neuronal plasticity. In the adult brain, PNNs play a crucial role in the regulation of learning and cognitive processes. Neuropeptide Y (NPY) is involved in the regulation of many physiological functions, including learning and memory abilities, via activation of Y1 receptors (Y1Rs). Here we demonstrated that the conditional depletion of the gene encoding the Y1R for NPY in adult forebrain excitatory neurons (Npy1rrfb mutant mice), induces a significant slowdown in spatial learning, which is associated with a robust intensification of PNN expression and an increase in the number of c-Fos expressing cells in the cornus ammonis 1 (CA1) of the dorsal hippocampus. Importantly, the enzymatic digestion of PNNs in CA1 normalizes c-Fos activity and completely rescues learning abilities of Npy1rrfb mice. These data highlight a previously unknown functional link between NPY-Y1R transmission and PNNs, which may play a role in the control of dorsal hippocampal excitability and related cognitive functions.
Collapse
Affiliation(s)
- Ilaria Bertocchi
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy
| | - Paolo Mele
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy
| | - Giuliano Ferrero
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy
| | - Alessandra Oberto
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy
| | - Daniela Carulli
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy; Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, Netherlands
| | - Carola Eva
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy.
| |
Collapse
|
12
|
Bioinformatics and experimental studies on the structural roles of a surface-exposed α-helix at the C-terminal domain of Chondroitinase ABC I. Int J Biol Macromol 2020; 163:1572-1578. [PMID: 32791283 DOI: 10.1016/j.ijbiomac.2020.07.165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 11/20/2022]
Abstract
A series of single and double mutants generated on residues of a surfaced-exposed helix at the C-terminal domain of chondroitinase ABC I (cABC I) from proteus vulgaris. M886A, G887E, and their respective double mutant, MA/GE were inspired by the sequence of a similar helix segment in 30S ribosomal protein S1. Additionally, M889I, Q891K, and the corresponding double mutant, MI/QK, were made regarding the sequence of a similar helix in chondroitin lyase from Proteus mirabilis. Circular dichroism spectra in the far-UV region, demonstrate that the ordered structure of wild-type (WT), and double mutants are the same; however, the helicity of the ordered structures in MI/QK is higher than that of the WT enzyme. When compared with the single mutants, the double mutants showed higher activity, and that the activity of MI/QK is higher than that of the WT enzyme. Heat-induced denaturation experiments showed that the stability of the tertiary structure of double mutants at moderate temperatures is higher compared with the WT, and single mutants. It concluded that this helix can be considered as one of the hot spots region that can be more manipulated to obtain improved variants of cABC I.
Collapse
|
13
|
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
|
14
|
Urban MW, Ghosh B, Block CG, Charsar BA, Smith GM, Wright MC, Li S, Lepore AC. Protein Tyrosine Phosphatase σ Inhibitory Peptide Promotes Recovery of Diaphragm Function and Sprouting of Bulbospinal Respiratory Axons after Cervical Spinal Cord Injury. J Neurotrauma 2019; 37:572-579. [PMID: 31392919 DOI: 10.1089/neu.2019.6586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Damage to respiratory neural circuitry and consequent loss of diaphragm function is a major cause of morbidity and mortality after cervical spinal cord injury (SCI). Upon SCI, inspiratory signals originating in the medullary rostral ventral respiratory group (rVRG) become disrupted from their phrenic motor neuron (PhMN) targets, resulting in diaphragm paralysis. Limited growth of both damaged and spared axon populations occurs after central nervous system trauma attributed, in part, to expression of various growth inhibitory molecules, some that act through direct interaction with the protein tyrosine phosphatase sigma (PTPσ) receptor located on axons. In the rat model of C2 hemisection SCI, we aimed to block PTPσ signaling to investigate potential mechanisms of axon plasticity and respiratory recovery using a small molecule peptide mimetic that inhibits PTPσ. The peptide was soaked into a biocompatible gelfoam and placed directly over the injury site immediately after hemisection and replaced with a freshly soaked piece 1 week post-SCI. At 8 weeks post-hemisection, PTPσ peptide significantly improved ipsilateral hemidiaphragm function, as assessed in vivo with electromyography recordings. PTPσ peptide did not promote regeneration of axotomized rVRG fibers originating in ipsilateral medulla, as assessed by tracing after adeno-associated virus serotype 2/mCherry injection into the rVRG. Conversely, PTPσ peptide stimulated robust sprouting of contralateral-originating rVRG fibers and serotonergic axons within the PhMN pool ipsilateral to hemisection. Further, relesion through the hemisection did not compromise diaphragm recovery, suggesting that PTPσ peptide-induced restoration of function was attributed to plasticity of spared axon pathways descending in contralateral spinal cord. These data demonstrate that inhibition of PTPσ signaling can promote significant recovery of diaphragm function after SCI by stimulating plasticity of critical axon populations spared by the injury and consequently enhancing descending excitatory input to PhMNs.
Collapse
Affiliation(s)
- Mark W Urban
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Biswarup Ghosh
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Cole G Block
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Brittany A Charsar
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - George M Smith
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Megan C Wright
- Department of Biology, Arcadia University, Glenside, Pennsylvania
| | - Shuxin Li
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Song YH, Agrawal NK, Griffin JM, Schmidt CE. Recent advances in nanotherapeutic strategies for spinal cord injury repair. Adv Drug Deliv Rev 2019; 148:38-59. [PMID: 30582938 PMCID: PMC6959132 DOI: 10.1016/j.addr.2018.12.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/12/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a devastating and complicated condition with no cure available. The initial mechanical trauma is followed by a secondary injury characterized by inflammatory cell infiltration and inhibitory glial scar formation. Due to the limitations posed by the blood-spinal cord barrier, systemic delivery of therapeutics is challenging. Recent development of various nanoscale strategies provides exciting and promising new means of treating SCI by crossing the blood-spinal cord barrier and delivering therapeutics. As such, we discuss different nanomaterial fabrication methods and provide an overview of recent studies where nanomaterials were developed to modulate inflammatory signals, target inhibitory factors in the lesion, and promote axonal regeneration after SCI. We also review emerging areas of research such as optogenetics, immunotherapy and CRISPR-mediated genome editing where nanomaterials can provide synergistic effects in developing novel SCI therapy regimens, as well as current efforts and barriers to clinical translation of nanomaterials.
Collapse
Affiliation(s)
- Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nikunj K Agrawal
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jonathan M Griffin
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Bieler L, Vogl M, Kirchinger M, Urmann C, Riepl H, Bandtlow C, Klimaschewski L, Aigner L, Couillard-Despres S. The Prenylflavonoid ENDF1 Overrules Central Nervous System Growth Inhibitors and Facilitates Regeneration of DRG Neurons. Front Cell Neurosci 2019; 13:332. [PMID: 31396054 PMCID: PMC6668039 DOI: 10.3389/fncel.2019.00332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/04/2019] [Indexed: 01/04/2023] Open
Abstract
Restoration of neuronal connectivity after lesion of the central nervous system, such as spinal cord injury, is one of the biggest challenges in modern medicine. In particular, the accumulation of axon growth inhibitory factors at the site of injury constitutes a major obstacle to structural and thus functional repair. We previously investigated a group of prenylflavonoids derived from hops for their capacity to promote neuroregeneration. We identified a molecule called ENDF1 that was very potent to enhance regrowth and branching of neurites from dorsal root ganglion neurons in culture on growth promoting substrates. In the present study, we investigated ENDF1’s capacity to promote regeneration of rat dorsal root ganglion neurons in vitro in the presence of three main components of the extracellular matrix acting as axon growth inhibitors: Semaphorin 3A, Ephrin A4 and mixed chondroitin sulfate proteoglycans. We report that ENDF1 application significantly promoted the percentages of sensory neurons able to regrow their neurites regardless of the presence of those inhibitors, and this to an extent similar to the one obtained after NGF treatment. Moreover, ENDF1 strongly enhanced the total neurite length and the complexity of neurites extending from neurons challenged with axon growth inhibitors. Although the impact of NGF and ENDF1 on the regeneration of neurons was similar, the activity of ENDF1 was not mediated by signaling through the TrkA receptor, indicating that each molecule act through different signaling pathways. In addition, ENDF1 did not decrease the phosphorylation of cofilin, a downstream effector of the regeneration-associated RhoA/ROCK signaling pathway. Hence, ENDF1 is a potent pro-neuroregenerative factors that could help in identifying new efficient targets for regenerative therapies of the nervous system.
Collapse
Affiliation(s)
- Lara Bieler
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Michael Vogl
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Michael Kirchinger
- Organic-Analytical Chemistry, Weihenstephan-Triesdorf University of Applied Sciences, Straubing, Germany.,TUM Campus Straubing, Straubing, Germany
| | - Corinna Urmann
- Organic-Analytical Chemistry, Weihenstephan-Triesdorf University of Applied Sciences, Straubing, Germany.,TUM Campus Straubing, Straubing, Germany
| | - Herbert Riepl
- Organic-Analytical Chemistry, Weihenstephan-Triesdorf University of Applied Sciences, Straubing, Germany.,TUM Campus Straubing, Straubing, Germany
| | - Christine Bandtlow
- Division of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria
| | - Ludwig Aigner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
17
|
Hu HZ, Granger N, Pai SB, Bellamkonda RV, Jeffery ND. Therapeutic efficacy of microtube-embedded chondroitinase ABC in a canine clinical model of spinal cord injury. Brain 2019; 141:1017-1027. [PMID: 29444239 DOI: 10.1093/brain/awy007] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/28/2017] [Indexed: 01/26/2023] Open
Abstract
See Moon and Bradbury (doi:10.1093/brain/awy067) for a scientific commentary on this article.Many hundreds of thousands of people around the world are living with the long-term consequences of spinal cord injury and they need effective new therapies. Laboratory research in experimental animals has identified a large number of potentially translatable interventions but transition to the clinic is not straightforward. Further evidence of efficacy in more clinically-relevant lesions is required to gain sufficient confidence to commence human clinical trials. Of the many therapeutic candidates currently available, intraspinally applied chondroitinase ABC has particularly well documented efficacy in experimental animals. In this study we measured the effects of this intervention in a double-blinded randomized controlled trial in a cohort of dogs with naturally-occurring severe chronic spinal cord injuries that model the condition in humans. First, we collected baseline data on a series of outcomes: forelimb-hindlimb coordination (the prespecified primary outcome measure), skin sensitivity along the back, somatosensory evoked and transcranial magnetic motor evoked potentials and cystometry in 60 dogs with thoracolumbar lesions. Dogs were then randomized 1:1 to receive intraspinal injections of heat-stabilized, lipid microtube-embedded chondroitinase ABC or sham injections consisting of needle puncture of the skin. Outcome data were measured at 1, 3 and 6 months after intervention; skin sensitivity was also measured 24 h after injection (or sham). Forelimb-hindlimb coordination was affected by neither time nor chondroitinase treatment alone but there was a significant interaction between these variables such that coordination between forelimb and hindlimb stepping improved during the 6-month follow-up period in the chondroitinase-treated animals by a mean of 23%, but did not change in controls. Three dogs (10%) in the chondroitinase group also recovered the ability to ambulate without assistance. Sensitivity of the dorsal skin increased at 24 h after intervention in both groups but subsequently decreased to normal levels. Cystometry identified a non-significant improvement of bladder compliance at 1 month in the chondroitinase-injected dogs but this did not persist. There were no overall differences between groups in detection of sensory evoked potentials. Our results strongly support a beneficial effect of intraspinal injection of chondroitinase ABC on spinal cord function in this highly clinically-relevant model of chronic severe spinal cord injury. There was no evidence of long-term adverse effects associated with this intervention. We therefore conclude that this study provides strong evidence in support of initiation of clinical trials of chondroitinase ABC in humans with chronic spinal cord injury.
Collapse
Affiliation(s)
- Hilary Z Hu
- 1 College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames IA 50011, USA
| | - Nicolas Granger
- 2 Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, UK
| | - S Balakrishna Pai
- 3 Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Ravi V Bellamkonda
- 4 Department of Biomedical Engineering, Duke University Pratt School of Engineering, 101 Science Drive, Campus Box 90281, Durham NC 27708, USA
| | - Nick D Jeffery
- 1 College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames IA 50011, USA
| |
Collapse
|
18
|
Kramer J, Chirco KR, Lamba DA. Immunological Considerations for Retinal Stem Cell Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:99-119. [PMID: 31654387 DOI: 10.1007/978-3-030-28471-8_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is an increasing effort toward generating replacement cells for neuronal application due to the nonregenerative nature of these tissues. While much progress has been made toward developing methodologies to generate these cells, there have been limited improvements in functional restoration. Some of these are linked to the degenerative and often nonreceptive microenvironment that the new cells need to integrate into. In this chapter, we will focus on the status and role of the immune microenvironment of the retina during homeostasis and disease states. We will review changes in both innate and adaptive immunity as well as the role of immune rejection in stem cell replacement therapies. The chapter will end with a discussion of immune-modulatory strategies that have helped to ameliorate these effects and could potentially improve functional outcome for cell replacement therapies for the eye.
Collapse
Affiliation(s)
- Joshua Kramer
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Deepak A Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA. .,Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
19
|
Abstract
The retina is a very fine and layered neural tissue, which vitally depends on the preservation of cells, structure, connectivity and vasculature to maintain vision. There is an urgent need to find technical and biological solutions to major challenges associated with functional replacement of retinal cells. The major unmet challenges include generating sufficient numbers of specific cell types, achieving functional integration of transplanted cells, especially photoreceptors, and surgical delivery of retinal cells or tissue without triggering immune responses, inflammation and/or remodeling. The advances of regenerative medicine enabled generation of three-dimensional tissues (organoids), partially recreating the anatomical structure, biological complexity and physiology of several tissues, which are important targets for stem cell replacement therapies. Derivation of retinal tissue in a dish creates new opportunities for cell replacement therapies of blindness and addresses the need to preserve retinal architecture to restore vision. Retinal cell therapies aimed at preserving and improving vision have achieved many improvements in the past ten years. Retinal organoid technologies provide a number of solutions to technical and biological challenges associated with functional replacement of retinal cells to achieve long-term vision restoration. Our review summarizes the progress in cell therapies of retina, with focus on human pluripotent stem cell-derived retinal tissue, and critically evaluates the potential of retinal organoid approaches to solve a major unmet clinical need—retinal repair and vision restoration in conditions caused by retinal degeneration and traumatic ocular injuries. We also analyze obstacles in commercialization of retinal organoid technology for clinical application.
Collapse
|
20
|
Nori S, Khazaei M, Ahuja CS, Yokota K, Ahlfors JE, Liu Y, Wang J, Shibata S, Chio J, Hettiaratchi MH, Führmann T, Shoichet MS, Fehlings MG. Human Oligodendrogenic Neural Progenitor Cells Delivered with Chondroitinase ABC Facilitate Functional Repair of Chronic Spinal Cord Injury. Stem Cell Reports 2018; 11:1433-1448. [PMID: 30472009 PMCID: PMC6294173 DOI: 10.1016/j.stemcr.2018.10.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 01/09/2023] Open
Abstract
Treatment of chronic spinal cord injury (SCI) is challenging due to cell loss, cyst formation, and the glial scar. Previously, we reported on the therapeutic potential of a neural progenitor cell (NPC) and chondroitinase ABC (ChABC) combinatorial therapy for chronic SCI. However, the source of NPCs and delivery system required for ChABC remained barriers to clinical application. Here, we investigated directly reprogrammed human NPCs biased toward an oligodendrogenic fate (oNPCs) in combination with sustained delivery of ChABC using an innovative affinity release strategy in a crosslinked methylcellulose biomaterial for the treatment of chronic SCI in an immunodeficient rat model. This combinatorial therapy increased long-term survival of oNPCs around the lesion epicenter, facilitated greater oligodendrocyte differentiation, remyelination of the spared axons by engrafted oNPCs, enhanced synaptic connectivity with anterior horn cells and neurobehavioral recovery. This combinatorial therapy is a promising strategy to regenerate the chronically injured spinal cord. Sustained biomaterial delivery of ChABC successfully degraded CSPGs XMC-ChABC promoted differentiation of oNPCs to more oligodendrocytes XMC-ChABC increased the long-term survival and integration of grafted oNPCs XMC-ChABC and oNPC combinatorial therapy is a promising treatment for chronic SCI
Collapse
Affiliation(s)
- Satoshi Nori
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinju-ku, Tokyo 160-8582, Japan
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Christopher S Ahuja
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Kazuya Yokota
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jan-Eric Ahlfors
- New World Laboratories Inc., 500 Boulevard Cartier Quest, Laval, QC H7V 5B7, Canada
| | - Yang Liu
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Jian Wang
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, 35 Shinanomachi, Shinju-ku, Tokyo 160-8582, Japan
| | - Jonathon Chio
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Marian H Hettiaratchi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada
| | - Tobias Führmann
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON M5S 3H6, Canada; Institute of Biomaterials & Biomedical Engineering, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Surgery and Spinal Program, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Surgery, Division of Anatomy, Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada.
| |
Collapse
|
21
|
Paylor JW, Wendlandt E, Freeman TS, Greba Q, Marks WN, Howland JG, Winship IR. Impaired Cognitive Function after Perineuronal Net Degradation in the Medial Prefrontal Cortex. eNeuro 2018; 5:ENEURO.0253-18.2018. [PMID: 30627657 PMCID: PMC6325561 DOI: 10.1523/eneuro.0253-18.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/09/2018] [Accepted: 11/29/2018] [Indexed: 11/21/2022] Open
Abstract
Perineuronal nets (PNNs) are highly organized components of the extracellular matrix that surround a subset of mature neurons in the CNS. These structures play a critical role in regulating neuronal plasticity, particularly during neurodevelopment. Consistent with this role, their presence is associated with functional and structural stability of the neurons they ensheath. A loss of PNNs in the prefrontal cortex (PFC) has been suggested to contribute to cognitive impairment in disorders such as schizophrenia. However, the direct consequences of PNN loss in medial PFC (mPFC) on cognition has not been demonstrated. Here, we examined behavior after disruption of PNNs in mPFC of Long-Evans rats following injection of the enzyme chondroitinase ABC (ChABC). Our data show that ChABC-treated animals were impaired on tests of object oddity perception. Performance in the cross-modal object recognition (CMOR) task was not significantly different for ChABC-treated rats, although ChABC-treated rats were not able to perform above chance levels whereas control rats were. ChABC-treated animals were not significantly different from controls on tests of prepulse inhibition (PPI), set-shifting (SS), reversal learning, or tactile and visual object recognition memory. Posthumous immunohistochemistry confirmed significantly reduced PNNs in mPFC due to ChABC treatment. Moreover, PNN density in the mPFC predicted performance on the oddity task, where higher PNN density was associated with better performance. These findings suggest that PNN loss within the mPFC impairs some aspects of object oddity perception and recognition and that PNNs contribute to cognitive function in young adulthood.
Collapse
Affiliation(s)
- John W. Paylor
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Eszter Wendlandt
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Tara S. Freeman
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Quentin Greba
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E Canada
| | - Wendie N. Marks
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E Canada
| | - John G. Howland
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E Canada
| | - Ian R. Winship
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2R3 Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1 Canada
| |
Collapse
|
22
|
Kim Y, Kang H, Powathil G, Kim H, Trucu D, Lee W, Lawler S, Chaplain M. Role of extracellular matrix and microenvironment in regulation of tumor growth and LAR-mediated invasion in glioblastoma. PLoS One 2018; 13:e0204865. [PMID: 30286133 PMCID: PMC6171904 DOI: 10.1371/journal.pone.0204865] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
The cellular dispersion and therapeutic control of glioblastoma, the most aggressive type of primary brain cancer, depends critically on the migration patterns after surgery and intracellular responses of the individual cancer cells in response to external biochemical cues in the microenvironment. Recent studies have shown that miR-451 regulates downstream molecules including AMPK/CAB39/MARK and mTOR to determine the balance between rapid proliferation and invasion in response to metabolic stress in the harsh tumor microenvironment. Surgical removal of the main tumor is inevitably followed by recurrence of the tumor due to inaccessibility of dispersed tumor cells in normal brain tissue. In order to address this complex process of cell proliferation and invasion and its response to conventional treatment, we propose a mathematical model that analyzes the intracellular dynamics of the miR-451-AMPK- mTOR-cell cycle signaling pathway within a cell. The model identifies a key mechanism underlying the molecular switches between proliferative phase and migratory phase in response to metabolic stress in response to fluctuating glucose levels. We show how up- or down-regulation of components in these pathways affects the key cellular decision to infiltrate or proliferate in a complex microenvironment in the absence and presence of time delays and stochastic noise. Glycosylated chondroitin sulfate proteoglycans (CSPGs), a major component of the extracellular matrix (ECM) in the brain, contribute to the physical structure of the local brain microenvironment but also induce or inhibit glioma invasion by regulating the dynamics of the CSPG receptor LAR as well as the spatiotemporal activation status of resident astrocytes and tumor-associated microglia. Using a multi-scale mathematical model, we investigate a CSPG-induced switch between invasive and non-invasive tumors through the coordination of ECM-cell adhesion and dynamic changes in stromal cells. We show that the CSPG-rich microenvironment is associated with non-invasive tumor lesions through LAR-CSGAG binding while the absence of glycosylated CSPGs induce the critical glioma invasion. We illustrate how high molecular weight CSPGs can regulate the exodus of local reactive astrocytes from the main tumor lesion, leading to encapsulation of non-invasive tumor and inhibition of tumor invasion. These different CSPG conditions also change the spatial profiles of ramified and activated microglia. The complex distribution of CSPGs in the tumor microenvironment can determine the nonlinear invasion behaviors of glioma cells, which suggests the need for careful therapeutic strategies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
| | - Hyunji Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Gibin Powathil
- Department of Mathematics, Swansea University, Swansea, United Kingdom
| | - Hyeongi Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee, United Kingdom
| | - Wanho Lee
- National Institute for Mathematical Sciences, Daejeon, Republic of Korea
| | - Sean Lawler
- Department of neurosurgery, Brigham and Women’s Hospital & Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mark Chaplain
- School of Mathematics and Statistics, Mathematical Institute, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
23
|
Patil N, Truong V, Holmberg MH, Lavoie NS, McCoy MR, Dutton JR, Holmberg EG, Parr AM. Safety and Efficacy of Rose Bengal Derivatives for Glial Scar Ablation in Chronic Spinal Cord Injury. J Neurotrauma 2018; 35:1745-1754. [PMID: 29373946 PMCID: PMC6033306 DOI: 10.1089/neu.2017.5398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There are no effective therapies available currently to ameliorate loss of function for patients with spinal cord injuries (SCIs). In addition, proposed treatments that demonstrated functional recovery in animal models of acute SCI have failed almost invariably when applied to chronic injury models. Glial scar formation in chronic injury is a likely contributor to limitation on regeneration. We have removed existing scar tissue in chronically contused rat spinal cord using a rose Bengal-based photo ablation approach. In this study, we compared two chemically modified rose bengal derivatives to unmodified rose bengal, both confirming and expanding on our previously published report. Rats were treated with unmodified rose bengal (RB1) or rose bengal modified with hydrocarbon (RB2) or polyethylene glycol (RB3), to determine the effects on scar components and spared tissue post-treatment. Our results showed that RB1 was more efficacious than RB2, while still maintaining minimal collateral effects on spared tissue. RB3 was not taken up by the cells, likely because of its size, and therefore had no effect. Treatment with RB1 also resulted in an increase in serotonin eight days post-treatment in chronically injured spinal cords. Thus, we suggest that unmodified rose Bengal is a potent candidate agent for the development of a therapeutic strategy for scar ablation in chronic SCI.
Collapse
Affiliation(s)
- Nandadevi Patil
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Vincent Truong
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Mackenzie H. Holmberg
- Department of Chemistry, University of Alaska Anchorage, Anchorage, Alaska
- University of Washington School of Medicine, Seattle, Washington
| | - Nicolas S. Lavoie
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Mark R. McCoy
- Department of Chemistry, University of Alaska Anchorage, Anchorage, Alaska
| | - James R. Dutton
- Department of Genetics, Cell Biology and Development, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Eric G. Holmberg
- Department of Chemistry, University of Alaska Anchorage, Anchorage, Alaska
| | - Ann M. Parr
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
24
|
Gómez RM, Sánchez MY, Portela-Lomba M, Ghotme K, Barreto GE, Sierra J, Moreno-Flores MT. Cell therapy for spinal cord injury with olfactory ensheathing glia cells (OECs). Glia 2018; 66:1267-1301. [PMID: 29330870 DOI: 10.1002/glia.23282] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 01/18/2023]
Abstract
The prospects of achieving regeneration in the central nervous system (CNS) have changed, as most recent findings indicate that several species, including humans, can produce neurons in adulthood. Studies targeting this property may be considered as potential therapeutic strategies to respond to injury or the effects of demyelinating diseases in the CNS. While CNS trauma may interrupt the axonal tracts that connect neurons with their targets, some neurons remain alive, as seen in optic nerve and spinal cord (SC) injuries (SCIs). The devastating consequences of SCIs are due to the immediate and significant disruption of the ascending and descending spinal pathways, which result in varying degrees of motor and sensory impairment. Recent therapeutic studies for SCI have focused on cell transplantation in animal models, using cells capable of inducing axon regeneration like Schwann cells (SchCs), astrocytes, genetically modified fibroblasts and olfactory ensheathing glia cells (OECs). Nevertheless, and despite the improvements in such cell-based therapeutic strategies, there is still little information regarding the mechanisms underlying the success of transplantation and regarding any secondary effects. Therefore, further studies are needed to clarify these issues. In this review, we highlight the properties of OECs that make them suitable to achieve neuroplasticity/neuroregeneration in SCI. OECs can interact with the glial scar, stimulate angiogenesis, axon outgrowth and remyelination, improving functional outcomes following lesion. Furthermore, we present evidence of the utility of cell therapy with OECs to treat SCI, both from animal models and clinical studies performed on SCI patients, providing promising results for future treatments.
Collapse
Affiliation(s)
- Rosa M Gómez
- Fundación de Neuroregeneración en Colombia, Grupo de investigación NeuroRec, Bogota D.C, Colombia
| | - Magdy Y Sánchez
- Fundación de Neuroregeneración en Colombia, Grupo de investigación NeuroRec, Bogota D.C, Colombia.,Maestría en Neurociencias, Universidad Nacional de Colombia, Bogota D.C, Colombia
| | - Maria Portela-Lomba
- Facultad de CC Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Kemel Ghotme
- Facultad de Medicina, Universidad de la Sabana, Chía, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota D.C, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Javier Sierra
- Facultad de CC Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | | |
Collapse
|
25
|
Kataria H, Alizadeh A, Shahriary GM, Saboktakin Rizi S, Henrie R, Santhosh KT, Thliveris JA, Karimi-Abdolrezaee S. Neuregulin-1 promotes remyelination and fosters a pro-regenerative inflammatory response in focal demyelinating lesions of the spinal cord. Glia 2017; 66:538-561. [PMID: 29148104 DOI: 10.1002/glia.23264] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/26/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022]
Abstract
Oligodendroglial cell death and demyelination are hallmarks of neurotrauma and multiple sclerosis that cause axonal damage and functional impairments. Remyelination remains a challenge as the ability of endogenous precursor cells for oligodendrocyte replacement is hindered in the unfavorable milieu of demyelinating conditions. Here, in a rat model of lysolecithin lysophosphatidyl-choline (LPC)-induced focal demyelination, we report that Neuregulin-1 (Nrg-1), an important factor for oligodendrocytes and myelination, is dysregulated in demyelinating lesions and its bio-availability can promote oligodendrogenesis and remyelination. We delivered recombinant human Nrg-1β1 (rhNrg-1β1) intraspinally in the vicinity of LPC demyelinating lesion in a sustained manner using poly lactic-co-glycolic acid microcarriers. Availability of Nrg-1 promoted generation and maturation of new oligodendrocytes, and accelerated endogenous remyelination by both oligodendrocyte and Schwann cell populations in demyelinating foci. Importantly, Nrg-1 enhanced myelin thickness in newly remyelinated spinal cord axons. Our complementary in vitro studies also provided direct evidence that Nrg-1 significantly promotes maturation of new oligodendrocytes and facilitates their transition to a myelinating phenotype. Nrg-1 therapy remarkably attenuated the upregulated expression chondroitin sulfate proteoglycans (CSPGs) specific glycosaminoglycans in the extracellular matrix of demyelinating foci and promoted interleukin-10 (IL-10) production by immune cells. CSPGs and IL-10 are known to negatively and positively regulate remyelination, respectively. We found that Nrg-1 effects are mediated through ErbB2 and ErbB4 receptor activation. Our work provides novel evidence that dysregulated levels of Nrg-1 in demyelinating lesions of the spinal cord pose a challenge to endogenous remyelination, and appear to be an underlying cause of myelin thinning in newly remyelinated axons.
Collapse
Affiliation(s)
- Hardeep Kataria
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ghazaleh M Shahriary
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shekoofeh Saboktakin Rizi
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ryan Henrie
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kallivalappil T Santhosh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
26
|
Pakulska MM, Tator CH, Shoichet MS. Local delivery of chondroitinase ABC with or without stromal cell-derived factor 1α promotes functional repair in the injured rat spinal cord. Biomaterials 2017; 134:13-21. [DOI: 10.1016/j.biomaterials.2017.04.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 12/14/2022]
|
27
|
De Luca C, Papa M. Matrix Metalloproteinases, Neural Extracellular Matrix, and Central Nervous System Pathology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:167-202. [PMID: 28662822 DOI: 10.1016/bs.pmbts.2017.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The functionality and stability of the central nervous system (CNS) pabulum, called neural extracellular matrix (nECM), is paramount for the maintenance of a healthy network. The loosening or the damage of the scaffold disrupts synaptic transmission with the consequent imbalance of the neurotransmitters, reactive cells invasion, astrocytosis, new matrix deposition, digestion of the previous structure and ultimately, maladaptive plasticity with the loss of neuronal viability. nECM is constantly affected by CNS disorders, particularly in chronic modifying such as neurodegenerative disease, or in acute/subacute with chronic sequelae, like cerebrovascular and inflammatory pathology. Matrix metalloproteinases (MMPs) are the main interfering agent of nECM, guiding the balance of degradation and new deposition of proteins such as proteoglycans and glycoproteins, or glycosaminoglycans, such as hyaluronic acid. Activation of these enzymes is modulated by their physiologic inhibitors, the tissue inhibitors of MMPs or via other proteases inhibitors, as well as genetic or epigenetic up- or downregulation through molecular interaction or receptor activation. The appropriate understanding of the pathways underlying nECM modifications in CNS pathology is probably one of the pivotal future directions to identify the healthy brain network and subsequently design new therapies to interfere with the progression of the CNS disease and eventually find appropriate therapies.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Papa
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy; SYSBIO, Centre for Systems Biology, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
28
|
Wu JH, Li M, Liang Y, Lu T, Duan CY. Migration of Adipose-derived Mesenchymal Stem Cells Stably Expressing Chondroitinase ABC In vitro. Chin Med J (Engl) 2017; 129:1592-9. [PMID: 27364797 PMCID: PMC4931267 DOI: 10.4103/0366-6999.184464] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Several studies have revealed that adipose-derived mesenchymal stem cells (ADSCs) can be used as seed cells for the treatment of spinal cord injury (SCI). Chondroitinase ABC (ChABC) decomposes chondroitin sulfate proteoglycans in the glial scar that forms following SCI, allowing stem cells to penetrate through the scar and promote recovery of nerve function. This study aimed to establish ADSCs that stably express ChABC (ChABC-ADSCs) and evaluate the migratory capability of ChABC-ADSCs in vitro. METHODS ADSCs were obtained from Sprague-Dawley rats using secondary collagenase digestion. Their phenotypes were characterized using flow cytometry detection of cell surface antigens and their stem cell properties were confirmed by induction of differentiation. After successful culture, ADSCs were transfected with lentiviral vectors and ChABC-ADSCs were obtained. Proliferation curves of ChABC-ADSCs were determined using the Cell Counting Kit-8 method, ChABC expression was verified using Western blotting, and the migration of ChABC-ADSCs was analyzed using the transwell assay. RESULTS Secondary collagenase digestion increased the isolation efficiency of primary ADSCs. Following transfection using lentiviral vectors, the proliferation of ChABC-ADSCs was reduced in comparison with control ADSCs at 48 h (P < 0.05). And the level of ChABC expression in the ChABC-ADSC group was significantly higher than that of the ADSC group (P < 0.05). Moreover, ChABC-ADSC migration in matrigel was significantly enhanced in comparison with the control (P < 0.05). CONCLUSIONS Secondary collagenase digestion can be used to effectively isolate ADSCs. ChABC-ADSCs constructed using lentiviral vector transfection stably express ChABC, and ChABC expression significantly enhances the migratory capacity of ADSCs.
Collapse
Affiliation(s)
- Jian-Huang Wu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Miao Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Liang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tao Lu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chun-Yue Duan
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
29
|
Transplantation of hUC-MSCs seeded collagen scaffolds reduces scar formation and promotes functional recovery in canines with chronic spinal cord injury. Sci Rep 2017; 7:43559. [PMID: 28262732 PMCID: PMC5337930 DOI: 10.1038/srep43559] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/25/2017] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) can lead to locomotor deficits, and the repair of chronic SCI is considered one of the most challenging clinical problems. Although extensive studies have evaluated treatments for acute SCI in small animals, comparatively fewer studies have been conducted on large-animal SCI in the chronic phase, which is more clinically relevant. Here, we used a collagen-based biomaterial, named the NeuroRegen scaffold, loaded with human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in a canine chronic SCI model. To generate chronic SCI, the T8 spinal cord segment was removed by complete transection of the spinal cord. Two months later, glial scar tissue was removed and a NeuroRegen scaffold was transplanted into the lesion area. Functionalized NeuroRegen scaffold implantation promoted both locomotor recovery and endogenous neurogenesis in the lesion area. Moreover, some newly generated neurons successfully matured into 5-HT-positive neurons at 1 year post-injury. In addition, many regenerated axon fibers in the lesion area exhibited remyelination and synapse formation at 1 year post-injury in the functionalized NeuroRegen scaffold group. In conclusion, the NeuroRegen scaffold functionalized with hUC-MSCs is a promising potential therapeutic approach to chronic SCI that promotes neuronal regeneration, reduces glial scar formation, and ultimately improves locomotor recovery.
Collapse
|
30
|
Hackett AR, Lee JK. Understanding the NG2 Glial Scar after Spinal Cord Injury. Front Neurol 2016; 7:199. [PMID: 27895617 PMCID: PMC5108923 DOI: 10.3389/fneur.2016.00199] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/31/2016] [Indexed: 01/05/2023] Open
Abstract
NG2 cells, also known as oligodendrocyte progenitor cells, are located throughout the central nervous system and serve as a pool of progenitors to differentiate into oligodendrocytes. In response to spinal cord injury (SCI), NG2 cells increase their proliferation and differentiation into remyelinating oligodendrocytes. While astrocytes are typically associated with being the major cell type in the glial scar, many NG2 cells also accumulate within the glial scar but their function remains poorly understood. Similar to astrocytes, these cells hypertrophy, upregulate expression of chondroitin sulfate proteoglycans, inhibit axon regeneration, contribute to the glial-fibrotic scar border, and some even differentiate into astrocytes. Whether NG2 cells also have a role in other astrocyte functions, such as preventing the spread of infiltrating leukocytes and expression of inflammatory cytokines, is not yet known. Thus, NG2 cells are not only important for remyelination after SCI but are also a major component of the glial scar with functions that overlap with astrocytes in this region. In this review, we describe the signaling pathways important for the proliferation and differentiation of NG2 cells, as well as the role of NG2 cells in scar formation and tissue repair.
Collapse
Affiliation(s)
- Amber R. Hackett
- Miami Project to Cure Paralysis, The Neuroscience Graduate Program, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jae K. Lee
- Miami Project to Cure Paralysis, The Neuroscience Graduate Program, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
31
|
Park HW, Jeon HJ, Chang MS. Vascular endothelial growth factor enhances axonal outgrowth in organotypic spinal cord slices via vascular endothelial growth factor receptor 1 and 2. Tissue Eng Regen Med 2016; 13:601-609. [PMID: 30603441 DOI: 10.1007/s13770-016-0051-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/13/2016] [Accepted: 06/01/2016] [Indexed: 01/26/2023] Open
Abstract
Enhancing adult nerve regeneration is a potential therapeutic strategy for treating spinal cord injury. Vascular endothelial growth factor (VEGF) is a major contributor to angiogenesis, which can reduce the spinal cord injury by inhibiting the inflammation and improve recovery after spinal cord injury. We have previously demonstrated that exogenous VEGF has neurotrophic effects on injured spinal nerves in organotypic spinal cord slice cultures. However, the mechanisms underlying the neurite growth by exogenous VEGF remain to be explored in spinal cord. In this study, we found out that exogenous VEGF mediated axonal outgrowth through VEGF receptor 1 (VEGFR1) and VEGFR2, both of which were expressed on organotypic spinal cord slices. Although VEGFR1 and VEGFR2 were constitutively expressed in some cells of control spinal cord slices, VEGF treatment upregulated expression of VEGFR1 and VEGFR2. Both VEGFR1 and VEGFR2 were expressed in neuronal cells as well as glial cells of organotypic spinal cord slices. We also observed that VEGF-induced axonal outgrowth was attenuated by a specific mitogen-activated protein kinase (MAPK) inhibitor PD98059 and a specific phosphoinositide 3-kinase (PI3K) inhibitor wortmannin. Thus, these findings suggest that these MAPK and PI3K pathways have important roles in regulating VEGF-induced axonal outgrowth in the postnatal spinal cord.
Collapse
Affiliation(s)
- Hwan-Woo Park
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 2Department of Cell Biology, College of Medicine, Konyang University, Daejeon, Korea
| | - Hyo-Jin Jeon
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Mi-Sook Chang
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 3Neuroscience Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
32
|
Tedcastle A, Illingworth S, Brown A, Seymour LW, Fisher KD. Actin-resistant DNAse I Expression From Oncolytic Adenovirus Enadenotucirev Enhances Its Intratumoral Spread and Reduces Tumor Growth. Mol Ther 2016; 24:796-804. [PMID: 26708004 PMCID: PMC4886935 DOI: 10.1038/mt.2015.233] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 12/17/2015] [Indexed: 12/27/2022] Open
Abstract
Spread of oncolytic viruses through tumor tissue is essential to effective virotherapy. Interstitial matrix is thought to be a significant barrier to virus particle convection between "islands" of tumor cells. One way to address this is to encode matrix-degrading enzymes within oncolytic viruses, for secretion from infected cells. To test the hypothesis that extracellular DNA provides an important barrier, we assessed the ability of DNase to promote virus spread. Nonreplicating Ad5 vectors expressing actin-resistant DNase (aDNAse I), proteinase K (PK), hyaluronidase (rhPH20), and chondroitinase ABC (CABC) were injected into established DLD human colorectal adenocarcinoma xenografts, transcomplemented with a replicating Ad5 virus. Each enzyme improved oncolysis by the replicating adenovirus, with no evidence of tumor cells being shed into the bloodstream. aDNAse I and rhPH20 hyaluronidase were then cloned into conditionally-replicating group B adenovirus, Enadenotucirev (EnAd). EnAd encoding each enzyme showed significantly better antitumor efficacy than the parental virus, with the aDNAse I-expressing virus showing improved spread. Both DNase and hyaluronidase activity was still measurable 32 days postinfection. This is the first time that extracellular DNA has been implicated as a barrier for interstitial virus spread, and suggests that oncolytic viruses expressing aDNAse I may be promising candidates for clinical translation.
Collapse
Affiliation(s)
| | | | | | | | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Rost S, Akyüz N, Martinovic T, Huckhagel T, Jakovcevski I, Schachner M. Germline ablation of dermatan-4O-sulfotransferase1 reduces regeneration after mouse spinal cord injury. Neuroscience 2016; 312:74-85. [PMID: 26586562 DOI: 10.1016/j.neuroscience.2015.11.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/02/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023]
Abstract
Chondroitin/dermatan sulfate proteoglycans (CSPGs/DSPGs) are major components of the extracellular matrix. Their expression is generally upregulated after injuries to the adult mammalian central nervous system, which is known for its low ability to restore function after injury. Several studies support the view that CSPGs inhibit regeneration after injury, whereas the functions of DSPGs in injury paradigms are less certain. To characterize the functions of DSPGs in the presence of CSPGs, we studied young adult dermatan-4O-sulfotransferase1-deficient (Chst14(-/-)) mice, which express chondroitin sulfates (CSs), but not dermatan sulfates (DSs), to characterize the functional outcome after severe compression injury of the spinal cord. In comparison to their wild-type (Chst14(+/+)) littermates, regeneration was reduced in Chst14(-/-) mice. No differences between genotypes were seen in the size of spinal cords, numbers of microglia and astrocytes neither in intact nor injured spinal cords after injury. Monoaminergic innervation and re-innervation of the spinal cord caudal to the lesion site as well as expression levels of glial fibrillary acidic protein (GFAP) and myelin basic protein (MBP) were similar in both genotypes, independent of whether they were injured and examined 6weeks after injury or not injured. These results suggest that, in contrast to CSPGs, DSPGs, being the products of Chst14 enzymatic activity, promote regeneration after injury of the adult mouse central nervous system.
Collapse
Affiliation(s)
- S Rost
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - N Akyüz
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - T Martinovic
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany; Institute of Histology and Embryology, School of Medicine, University of Belgrade, Višegradska 26, Belgrade, Serbia
| | - T Huckhagel
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - I Jakovcevski
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany; Experimental Neurophysiology, University Hospital Cologne, Joseph-Stelzmann-Str. 9, D-50931 Köln, Germany; German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, D-53175 Bonn, Germany.
| | - M Schachner
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany; Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, PR China; Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
34
|
Neuron-Glia Interactions in Neural Plasticity: Contributions of Neural Extracellular Matrix and Perineuronal Nets. Neural Plast 2016; 2016:5214961. [PMID: 26881114 PMCID: PMC4736403 DOI: 10.1155/2016/5214961] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/10/2015] [Indexed: 11/17/2022] Open
Abstract
Synapses are specialized structures that mediate rapid and efficient signal transmission between neurons and are surrounded by glial cells. Astrocytes develop an intimate association with synapses in the central nervous system (CNS) and contribute to the regulation of ion and neurotransmitter concentrations. Together with neurons, they shape intercellular space to provide a stable milieu for neuronal activity. Extracellular matrix (ECM) components are synthesized by both neurons and astrocytes and play an important role in the formation, maintenance, and function of synapses in the CNS. The components of the ECM have been detected near glial processes, which abut onto the CNS synaptic unit, where they are part of the specialized macromolecular assemblies, termed perineuronal nets (PNNs). PNNs have originally been discovered by Golgi and represent a molecular scaffold deposited in the interface between the astrocyte and subsets of neurons in the vicinity of the synapse. Recent reports strongly suggest that PNNs are tightly involved in the regulation of synaptic plasticity. Moreover, several studies have implicated PNNs and the neural ECM in neuropsychiatric diseases. Here, we highlight current concepts relating to neural ECM and PNNs and describe an in vitro approach that allows for the investigation of ECM functions for synaptogenesis.
Collapse
|
35
|
Siebert JR, Eade AM, Osterhout DJ. Biomaterial Approaches to Enhancing Neurorestoration after Spinal Cord Injury: Strategies for Overcoming Inherent Biological Obstacles. BIOMED RESEARCH INTERNATIONAL 2015; 2015:752572. [PMID: 26491685 PMCID: PMC4600545 DOI: 10.1155/2015/752572] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 01/14/2023]
Abstract
While advances in technology and medicine have improved both longevity and quality of life in patients living with a spinal cord injury, restoration of full motor function is not often achieved. This is due to the failure of repair and regeneration of neuronal connections in the spinal cord after injury. In this review, the complicated nature of spinal cord injury is described, noting the numerous cellular and molecular events that occur in the central nervous system following a traumatic lesion. In short, postinjury tissue changes create a complex and dynamic environment that is highly inhibitory to the process of neural regeneration. Strategies for repair are outlined with a particular focus on the important role of biomaterials in designing a therapeutic treatment that can overcome this inhibitory environment. The importance of considering the inherent biological response of the central nervous system to both injury and subsequent therapeutic interventions is highlighted as a key consideration for all attempts at improving functional recovery.
Collapse
Affiliation(s)
- Justin R. Siebert
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA 15601, USA
| | - Amber M. Eade
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA 15601, USA
| | - Donna J. Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
36
|
Cheng CH, Lin CT, Lee MJ, Tsai MJ, Huang WH, Huang MC, Lin YL, Chen CJ, Huang WC, Cheng H. Local Delivery of High-Dose Chondroitinase ABC in the Sub-Acute Stage Promotes Axonal Outgrowth and Functional Recovery after Complete Spinal Cord Transection. PLoS One 2015; 10:e0138705. [PMID: 26393921 PMCID: PMC4579094 DOI: 10.1371/journal.pone.0138705] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 09/02/2015] [Indexed: 01/11/2023] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are glial scar-associated molecules considered axonal regeneration inhibitors and can be digested by chondroitinase ABC (ChABC) to promote axonal regeneration after spinal cord injury (SCI). We previously demonstrated that intrathecal delivery of low-dose ChABC (1 U) in the acute stage of SCI promoted axonal regrowth and functional recovery. In this study, high-dose ChABC (50 U) introduced via intrathecal delivery induced subarachnoid hemorrhage and death within 48 h. However, most SCI patients are treated in the sub-acute or chronic stages, when the dense glial scar has formed and is minimally digested by intrathecal delivery of ChABC at the injury site. The present study investigated whether intraparenchymal delivery of ChABC in the sub-acute stage of complete spinal cord transection would promote axonal outgrowth and improve functional recovery. We observed no functional recovery following the low-dose ChABC (1 U or 5 U) treatments. Furthermore, animals treated with high-dose ChABC (50 U or 100 U) showed decreased CSPGs levels. The extent and area of the lesion were also dramatically decreased after ChABC treatment. The outgrowth of the regenerating axons was significantly increased, and some partially crossed the lesion site in the ChABC-treated groups. In addition, retrograde Fluoro-Gold (FG) labeling showed that the outgrowing axons could cross the lesion site and reach several brain stem nuclei involved in sensory and motor functions. The Basso, Beattie and Bresnahan (BBB) open field locomotor scores revealed that the ChABC treatment significantly improved functional recovery compared to the control group at eight weeks after treatment. Our study demonstrates that high-dose ChABC treatment in the sub-acute stage of SCI effectively improves glial scar digestion by reducing the lesion size and increasing axonal regrowth to the related functional nuclei, which promotes locomotor recovery. Thus, our results will aid in the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Chu-Hsun Cheng
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Te Lin
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Nursing, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Meng-Jen Lee
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Applied Chemistry, Chaoyang University of Technology, Taichung, Taiwan
| | - May-Jywan Tsai
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Hung Huang
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Chao Huang
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Lo Lin
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Jung Chen
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Cheng Huang
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Center for Neural Regeneration, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| | - Henrich Cheng
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Center for Neural Regeneration, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
37
|
Pires LR, Pêgo AP. Bridging the lesion-engineering a permissive substrate for nerve regeneration. Regen Biomater 2015; 2:203-14. [PMID: 26816642 PMCID: PMC4669012 DOI: 10.1093/rb/rbv012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/21/2015] [Accepted: 06/30/2015] [Indexed: 01/30/2023] Open
Abstract
Biomaterial-based strategies to restore connectivity after lesion at the spinal cord are focused on bridging the lesion and providing an favourable substrate and a path for axonal re-growth. Following spinal cord injury (SCI) a hostile environment for neuronal cell growth is established by the activation of multiple inhibitory mechanisms that hamper regeneration to occur. Implantable scaffolds can provide mechanical support and physical guidance for axon re-growth and, at the same time, contribute to alleviate the hostile environment by the in situ delivery of therapeutic molecules and/or relevant cells. Basic research on SCI has been contributing with the description of inhibitory mechanisms for regeneration as well as identifying drugs/molecules that can target inhibition. This knowledge is the background for the development of combined strategies with biomaterials. Additionally, scaffold design is significantly evolving. From the early simple hollow conduits, scaffolds with complex architectures that can modulate cell fate are currently being tested. A number of promising pre-clinical studies combining scaffolds, cells, drugs and/or nucleic acids are reported in the open literature. Overall, it is considered that to address the multi-factorial inhibitory environment of a SCI, a multifaceted therapeutic approach is imperative. The progress in the identification of molecules that target inhibition after SCI and its combination with scaffolds and/or cells are described and discussed in this review.
Collapse
Affiliation(s)
- Liliana R. Pires
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Faculdade de Engenharia—Universidade do Porto (FEUP), Porto, Portugal and
| | - Ana P. Pêgo
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Faculdade de Engenharia—Universidade do Porto (FEUP), Porto, Portugal and
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
38
|
Smith PD, Coulson-Thomas VJ, Foscarin S, Kwok JCF, Fawcett JW. "GAG-ing with the neuron": The role of glycosaminoglycan patterning in the central nervous system. Exp Neurol 2015; 274:100-14. [PMID: 26277685 DOI: 10.1016/j.expneurol.2015.08.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/17/2015] [Accepted: 08/06/2015] [Indexed: 01/17/2023]
Abstract
Proteoglycans (PGs) are a diverse family of proteins that consist of one or more glycosaminoglycan (GAG) chains, covalently linked to a core protein. PGs are major components of the extracellular matrix (ECM) and play critical roles in development, normal function and damage-response of the central nervous system (CNS). GAGs are classified based on their disaccharide subunits, into the following major groups: chondroitin sulfate (CS), heparan sulfate (HS), heparin (HEP), dermatan sulfate (DS), keratan sulfate (KS) and hyaluronic acid (HA). All except HA are modified by sulfation, giving GAG chains specific charged structures and binding properties. While significant neuroscience research has focused on the role of one PG family member, chondroitin sulfate proteoglycan (CSPG), there is ample evidence in support of a role for the other PGs in regulating CNS function in normal and pathological conditions. This review discusses the role of all the identified PG family members (CS, HS, HEP, DS, KS and HA) in normal CNS function and in the context of pathology. Understanding the pleiotropic roles of these molecules in the CNS may open the door to novel therapeutic strategies for a number of neurological conditions.
Collapse
Affiliation(s)
- Patrice D Smith
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK; Department of Neuroscience, Carleton University, Ottawa, ON, Canada.
| | - Vivien J Coulson-Thomas
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Simona Foscarin
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Jessica C F Kwok
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - James W Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK.
| |
Collapse
|
39
|
Bhattacharyya S, Zhang X, Feferman L, Johnson D, Tortella FC, Guizzetti M, Tobacman JK. Decline in arylsulfatase B and Increase in chondroitin 4-sulfotransferase combine to increase chondroitin 4-sulfate in traumatic brain injury. J Neurochem 2015; 134:728-39. [PMID: 25943740 DOI: 10.1111/jnc.13156] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/23/2015] [Accepted: 04/30/2015] [Indexed: 01/11/2023]
Abstract
In an established rat model of penetrating ballistic-like brain injury (PBBI), arylsulfatase B (ARSB; N-acetylgalactosamine 4-sulfatase) activity was significantly reduced at the ipsilateral site of injury, but unaffected at the contralateral site or in sham controls. In addition, the ARSB substrate chondroitin 4-sulfate (C4S) and total sulfated glycosaminoglycans increased. The mRNA expression of chondroitin 4-sulfotransferase 1 (C4ST1; CHST11) and the sulfotransferase activity rose at the ipsilateral site of injury (PBBI-I), indicating contributions from both increased production and reduced degradation to the accumulation of C4S. In cultured, fetal rat astrocytes, following scratch injury, the ARSB activity declined and the nuclear hypoxia inducible factor-1α increased significantly. In contrast, sulfotransferase activity and chondroitin 4-sulfotransferase expression increased following astrocyte exposure to TGF-β1, but not following scratch. These different pathways by which C4S increased in the cell preparations were both evident in the response to injury in the PBBI-I model. Hence, findings support effects of injury because of mechanical disruption inhibiting ARSB and to chemical mediation by TGF-β1 increasing CHST11 expression and sulfotransferase activity. The increase in C4S following traumatic brain injury is because of contributions from impaired degradation and enhanced synthesis of C4S which combine in the pathogenesis of the glial scar. This is the first report of how two mechanisms contribute to the increase in chondroitin 4-sulfate (C4S) in TBI. Following penetrating ballistic-like brain injury in a rat model and in the scratch model of injury in fetal rat astrocytes, Arylsulfatase B activity declined, leading to accumulation of C4S. TGF-β1 exposure increased expression of chondroitin 4-sulfotransferase. Hence, the increase in C4S in TBI is attributable to both impaired degradation and enhanced synthesis, combining in the pathogenesis of the glial scar.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Xiaolu Zhang
- Jesse Brown VA Medical Center, Chicago, Illinois, USA.,Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Leo Feferman
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - David Johnson
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Frank C Tortella
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Marina Guizzetti
- Jesse Brown VA Medical Center, Chicago, Illinois, USA.,Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA.,Oregon Health and Science University, Portland, Oregon, USA.,VA Portland Health Care System, Portland, Oregon, USA
| | - Joanne K Tobacman
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
40
|
Siddiqui AM, Khazaei M, Fehlings MG. Translating mechanisms of neuroprotection, regeneration, and repair to treatment of spinal cord injury. PROGRESS IN BRAIN RESEARCH 2015; 218:15-54. [PMID: 25890131 DOI: 10.1016/bs.pbr.2014.12.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the big challenges in neuroscience that remains to be understood is why the central nervous system is not able to regenerate to the extent that the peripheral nervous system does. This is especially problematic after traumatic injuries, like spinal cord injury (SCI), since the lack of regeneration leads to lifelong deficits and paralysis. Treatment of SCI has improved during the last several decades due to standardized protocols for emergency medical response teams and improved medical, surgical, and rehabilitative treatments. However, SCI continues to result in profound impairments for the individual. There are many processes that lead to the pathophysiology of SCI, such as ischemia, vascular disruption, neuroinflammation, oxidative stress, excitotoxicity, demyelination, and cell death. Current treatments include surgical decompression, hemodynamic control, and methylprednisolone. However, these early treatments are associated with modest functional recovery. Some treatments currently being investigated for use in SCI target neuroprotective (riluzole, minocycline, G-CSF, FGF-2, and polyethylene glycol) or neuroregenerative (chondroitinase ABC, self-assembling peptides, and rho inhibition) strategies, while many cell therapies (embryonic stem cells, neural stem cells, induced pluripotent stem cells, mesenchymal stromal cells, Schwann cells, olfactory ensheathing cells, and macrophages) have also shown promise. However, since SCI has multiple factors that determine the progress of the injury, a combinatorial therapeutic approach will most likely be required for the most effective treatment of SCI.
Collapse
Affiliation(s)
- Ahad M Siddiqui
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Mohamad Khazaei
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Critical Role of a Loop at C-Terminal Domain on the Conformational Stability and Catalytic Efficiency of Chondroitinase ABC I. Mol Biotechnol 2015; 57:727-34. [DOI: 10.1007/s12033-015-9864-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
42
|
Hippert C, Graca AB, Barber AC, West EL, Smith AJ, Ali RR, Pearson RA. Müller glia activation in response to inherited retinal degeneration is highly varied and disease-specific. PLoS One 2015; 10:e0120415. [PMID: 25793273 PMCID: PMC4368159 DOI: 10.1371/journal.pone.0120415] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/22/2015] [Indexed: 12/20/2022] Open
Abstract
Despite different aetiologies, most inherited retinal disorders culminate in photoreceptor loss, which induces concomitant changes in the neural retina, one of the most striking being reactive gliosis by Müller cells. It is typically assumed that photoreceptor loss leads to an upregulation of glial fibrilliary acidic protein (Gfap) and other intermediate filament proteins, together with other gliosis-related changes, including loss of integrity of the outer limiting membrane (OLM) and deposition of proteoglycans. However, this is based on a mix of both injury-induced and genetic causes of photoreceptor loss. There are very few longitudinal studies of gliosis in the retina and none comparing these changes across models over time. Here, we present a comprehensive spatiotemporal assessment of features of gliosis in the degenerating murine retina that involves Müller glia. Specifically, we assessed Gfap, vimentin and chondroitin sulphate proteoglycan (CSPG) levels and outer limiting membrane (OLM) integrity over time in four murine models of inherited photoreceptor degeneration that encompass a range of disease severities (Crb1rd8/rd8, Prph2+/Δ307, Rho-/-, Pde6brd1/rd1). These features underwent very different changes, depending upon the disease-causing mutation, and that these changes are not correlated with disease severity. Intermediate filament expression did indeed increase with disease progression in Crb1rd8/rd8 and Prph2+/Δ307, but decreased in the Prph2+/Δ307 and Pde6brd1/rd1 models. CSPG deposition usually, but not always, followed the trends in intermediate filament expression. The OLM adherens junctions underwent significant remodelling in all models, but with differences in the composition of the resulting junctions; in Rho-/- mice, the adherens junctions maintained the typical rod-Müller glia interactions, while in the Pde6brd1/rd1 model they formed predominantly between Müller cells in late stage of degeneration. Together, these results show that gliosis and its associated processes are variable and disease-dependent.
Collapse
Affiliation(s)
- Claire Hippert
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Anna B. Graca
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Amanda C. Barber
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Emma L. West
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Alexander J. Smith
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Robin R. Ali
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, City Road, London, EC1V 2PD, United Kingdom
| | - Rachael A. Pearson
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Xu CJ, Wang JL, Jin WL. The Neural Stem Cell Microenvironment: Focusing on Axon Guidance Molecules and Myelin-Associated Factors. J Mol Neurosci 2015; 56:887-897. [PMID: 25757451 DOI: 10.1007/s12031-015-0538-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/27/2015] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) could produce various cell phenotypes in the subventricular zone (SVZ) and dentate gyrus of the hippocampus in the central nervous system (CNS), where neurogenesis has been determined to occur. The extracellular microenvironment also influences the behaviors of NSCs during development and at CNS injury sites. Our previous study indicates that myelin, a component of the CNS, could regulate the differentiation of NSCs in vitro. Recent reports have implicated three myelin-derived inhibitors, NogoA, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp), as well as several axon guidance molecules as regulators of NSC survival, proliferation, migration, and differentiation. However, the molecular mechanisms underlying the behavior of NSCs are not fully understood. In this study, we summarize the current literature on the effects of different extrinsic factors on NSCs and discuss possible mechanisms, as well as future possible clinical applications.
Collapse
Affiliation(s)
- Chao-Jin Xu
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, University town, Cha Shan, Zhejiang, 325035, China.
| | - Jun-Ling Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China. .,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, 200240, China.
| |
Collapse
|
44
|
Xia Y, Yan Y, Xia H, Zhao T, Chu W, Hu S, Feng H, Lin J. Antisense vimentin cDNA combined with chondroitinase ABC promotes axon regeneration and functional recovery following spinal cord injury in rats. Neurosci Lett 2015; 590:74-9. [DOI: 10.1016/j.neulet.2015.01.073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 01/21/2015] [Accepted: 01/28/2015] [Indexed: 12/01/2022]
|
45
|
Mammalian target of rapamycin's distinct roles and effectiveness in promoting compensatory axonal sprouting in the injured CNS. J Neurosci 2015; 34:15347-55. [PMID: 25392502 DOI: 10.1523/jneurosci.1935-14.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) functions as a master sensor of nutrients and energy, and controls protein translation and cell growth. Deletion of phosphatase and tensin homolog (PTEN) in adult CNS neurons promotes regeneration of injured axons in an mTOR-dependent manner. However, others have demonstrated mTOR-independent axon regeneration in different cell types, raising the question of how broadly mTOR regulates axonal regrowth across different systems. Here we define the role of mTOR in promoting collateral sprouting of spared axons, a key axonal remodeling mechanism by which functions are recovered after CNS injury. Using pharmacological inhibition, we demonstrate that mTOR is dispensable for the robust spontaneous sprouting of corticospinal tract axons seen after pyramidotomy in postnatal mice. In contrast, moderate spontaneous axonal sprouting and induced-sprouting seen under different conditions in young adult mice (i.e., PTEN deletion or degradation of chondroitin proteoglycans; CSPGs) are both reduced upon mTOR inhibition. In addition, to further determine the potency of mTOR in promoting sprouting responses, we coinactivate PTEN and CSPGs, and demonstrate that this combination leads to an additive increase in axonal sprouting compared with single treatments. Our findings reveal a developmental switch in mTOR dependency for inducing axonal sprouting, and indicate that PTEN deletion in adult neurons neither recapitulates the regrowth program of postnatal animals, nor is sufficient to completely overcome an inhibitory environment. Accordingly, exploiting mTOR levels by targeting PTEN combined with CSPG degradation represents a promising strategy to promote extensive axonal plasticity in adult mammals.
Collapse
|
46
|
Jayakody SA, Gonzalez-Cordero A, Ali RR, Pearson RA. Cellular strategies for retinal repair by photoreceptor replacement. Prog Retin Eye Res 2015; 46:31-66. [PMID: 25660226 DOI: 10.1016/j.preteyeres.2015.01.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 01/13/2015] [Accepted: 01/19/2015] [Indexed: 02/08/2023]
Abstract
Loss of photoreceptors due to retinal degeneration is a major cause of blindness in the developed world. While no effective treatment is currently available, cell replacement therapy, using pluripotent stem cell-derived photoreceptor precursor cells, may be a feasible future treatment. Recent reports have demonstrated rescue of visual function following the transplantation of immature photoreceptors and we have seen major advances in our ability to generate transplantation-competent donor cells from stem cell sources. Moreover, we are beginning to realise the possibilities of using endogenous populations of cells from within the retina itself to mediate retinal repair. Here, we present a review of our current understanding of endogenous repair mechanisms together with recent progress in the use of both ocular and pluripotent stem cells for the treatment of photoreceptor loss. We consider how our understanding of retinal development has underpinned many of the recent major advances in translation and moved us closer to the goal of restoring vision by cellular means.
Collapse
Affiliation(s)
- Sujatha A Jayakody
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK
| | - Anai Gonzalez-Cordero
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK
| | - Robin R Ali
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK; NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, City Road, London EC1V 2PD, UK
| | - Rachael A Pearson
- Gene and Cell Therapy Group, Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath St, London EC1V 9EL, UK.
| |
Collapse
|
47
|
Vargas-Martínez F, Uvnäs-Moberg K, Petersson M, Olausson HA, Jiménez-Estrada I. Neuropeptides as neuroprotective agents: Oxytocin a forefront developmental player in the mammalian brain. Prog Neurobiol 2014; 123:37-78. [DOI: 10.1016/j.pneurobio.2014.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 02/07/2023]
|
48
|
Sivak WN, White JD, Bliley JM, Tien LW, Liao HT, Kaplan DL, Marra KG. Delivery of chondroitinase ABC and glial cell line-derived neurotrophic factor from silk fibroin conduits enhances peripheral nerve regeneration. J Tissue Eng Regen Med 2014; 11:733-742. [PMID: 25424415 DOI: 10.1002/term.1970] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/18/2014] [Accepted: 10/27/2014] [Indexed: 01/26/2023]
Abstract
Nerve conduits are a proven strategy for guiding axon regrowth following injury. This study compares degradable silk-trehalose films containing chondroitinase ABC (ChABC) and/or glial cell line-derived neurotrophic factor (GDNF) loaded within a silk fibroin-based nerve conduit in a rat sciatic nerve defect model. Four groups of silk conduits were prepared, with the following silk-trehalose films inserted into the conduit: (a) empty; (b) 1 µg GDNF; (3) 2 U ChABC; and (4) 1 µg GDNF/2 U ChABC. Drug release studies demonstrated 20% recovery of GDNF and ChABC at 6 weeks and 24 h, respectively. Six conduits of each type were implanted into 15 mm sciatic nerve defects in Lewis rats; conduits were explanted for histological analysis at 6 weeks. Tissues stained with Schwann cell S-100 antibody demonstrated an increased density of cells in both GDNF- and ChABC-treated groups compared to empty control conduits (p < 0.05). Conduits loaded with GDNF and ChABC also demonstrated higher levels of neuron-specific PGP 9.5 protein when compared to controls (p < 0.05). In this study we demonstrated a method to enhance Schwann cell migration and proliferation and also foster axonal regeneration when repairing peripheral nerve gap defects. Silk fibroin-based nerve conduits possess favourable mechanical and degradative properties and are further enhanced when loaded with ChABC and GDNF. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Wesley N Sivak
- Department of Plastic Surgery, University of Pittsburgh, PA, USA
| | - James D White
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | | | - Lee W Tien
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | - Han Tsung Liao
- Department of Plastic Surgery, University of Pittsburgh, PA, USA
- Department of Plastic and Reconstructive Surgery, Craniofacial Research Centre, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taiwan
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | - Kacey G Marra
- Department of Plastic Surgery, University of Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
49
|
Apara A, Goldberg JL. Molecular mechanisms of the suppression of axon regeneration by KLF transcription factors. Neural Regen Res 2014; 9:1418-21. [PMID: 25317150 PMCID: PMC4192940 DOI: 10.4103/1673-5374.139454] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 01/11/2023] Open
Abstract
Molecular mechanisms of the Krüppel-like family of transcription factors (KLFs) have been studied more in proliferating cells than in post-mitotic cells such as neurons. We recently found that KLFs regulate intrinsic axon growth ability in central nervous system (CNS) neurons including retinal ganglion cells, and hippocampal and cortical neurons. With at least 15 of 17 KLF family members expressed in neurons and at least 5 structurally unique subfamilies, it is important to determine how this complex family functions in neurons to regulate the intricate genetic programs of axon growth and regeneration. By characterizing the molecular mechanisms of the KLF family in the nervous system, including binding partners and gene targets, and comparing them to defined mechanisms defined outside the nervous system, we may better understand how KLFs regulate neurite growth and axon regeneration.
Collapse
Affiliation(s)
| | - Jeffrey L Goldberg
- Shiley Eye Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
50
|
Siebert JR, Conta Steencken A, Osterhout DJ. Chondroitin sulfate proteoglycans in the nervous system: inhibitors to repair. BIOMED RESEARCH INTERNATIONAL 2014; 2014:845323. [PMID: 25309928 PMCID: PMC4182688 DOI: 10.1155/2014/845323] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are widely expressed in the normal central nervous system, serving as guidance cues during development and modulating synaptic connections in the adult. With injury or disease, an increase in CSPG expression is commonly observed close to lesioned areas. However, these CSPG deposits form a substantial barrier to regeneration and are largely responsible for the inability to repair damage in the brain and spinal cord. This review discusses the role of CSPGs as inhibitors, the role of inflammation in stimulating CSPG expression near site of injury, and therapeutic strategies for overcoming the inhibitory effects of CSPGs and creating an environment conducive to nerve regeneration.
Collapse
Affiliation(s)
- Justin R. Siebert
- Lake Erie College of Osteopathic Medicine at Seton Hill, 20 Seton Hill Drive, Greensburg, PA 15601, USA
| | - Amanda Conta Steencken
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Donna J. Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|