1
|
Zhu W, Zhou Y, Guo L, Feng S. Biological function of sialic acid and sialylation in human health and disease. Cell Death Discov 2024; 10:415. [PMID: 39349440 PMCID: PMC11442784 DOI: 10.1038/s41420-024-02180-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024] Open
Abstract
Sialic acids are predominantly found at the terminal ends of glycoproteins and glycolipids and play key roles in cellular communication and function. The process of sialylation, a form of post-translational modification, involves the covalent attachment of sialic acid to the terminal residues of oligosaccharides and glycoproteins. This modification not only provides a layer of electrostatic repulsion to cells but also serves as a receptor for various biological signaling pathways. Sialylation is involved in several pathophysiological processes. Given its multifaceted involvement in cellular functions, sialylation presents a promising avenue for therapeutic intervention. Current studies are exploring agents that target sialic acid residues on sialoglycans or the sialylation process. These efforts are particularly focused on the fields of cancer therapy, stroke treatment, antiviral strategies, and therapies for central nervous system disorders. In this review, we aimed to summarize the biological functions of sialic acid and the process of sialylation, explore their roles in various pathophysiological contexts, and discuss their potential applications in the development of novel therapeutics.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yue Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linjuan Guo
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Shenghui Feng
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Granato V, Congiu L, Jakovcevski I, Kleene R, Schwindenhammer B, Fernandes L, Freitag S, Schachner M, Loers G. Mice Mutated in the First Fibronectin Domain of Adhesion Molecule L1 Show Brain Malformations and Behavioral Abnormalities. Biomolecules 2024; 14:468. [PMID: 38672483 PMCID: PMC11048097 DOI: 10.3390/biom14040468] [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: 02/08/2024] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The X-chromosome-linked cell adhesion molecule L1 (L1CAM), a glycoprotein mainly expressed by neurons in the central and peripheral nervous systems, has been implicated in many neural processes, including neuronal migration and survival, neuritogenesis, synapse formation, synaptic plasticity and regeneration. L1 consists of extracellular, transmembrane and cytoplasmic domains. Proteolytic cleavage of L1's extracellular and transmembrane domains by different proteases generates several L1 fragments with different functions. We found that myelin basic protein (MBP) cleaves L1's extracellular domain, leading to enhanced neuritogenesis and neuronal survival in vitro. To investigate in vivo the importance of the MBP-generated 70 kDa fragment (L1-70), we generated mice with an arginine to alanine substitution at position 687 (L1/687), thereby disrupting L1's MBP cleavage site and obliterating L1-70. Young adult L1/687 males showed normal anxiety and circadian rhythm activities but enhanced locomotion, while females showed altered social interactions. Older L1/687 males were impaired in motor coordination. Furthermore, L1/687 male and female mice had a larger hippocampus, with more neurons in the dentate gyrus and more proliferating cells in the subgranular layer, while the thickness of the corpus callosum and the size of lateral ventricles were normal. In summary, subtle mutant morphological changes result in subtle behavioral changes.
Collapse
Affiliation(s)
- Viviana Granato
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (V.G.); (L.C.); (R.K.); (S.F.)
| | - Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (V.G.); (L.C.); (R.K.); (S.F.)
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany; (I.J.); (B.S.)
- Department of Neuroanatomy and Molecular Brain Research, Institute of Anatomy, Ruhr-Universität Bochum, 44780 Bochum, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (V.G.); (L.C.); (R.K.); (S.F.)
| | - Benjamin Schwindenhammer
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany; (I.J.); (B.S.)
- Department of Neuroanatomy and Molecular Brain Research, Institute of Anatomy, Ruhr-Universität Bochum, 44780 Bochum, Germany
| | - Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (V.G.); (L.C.); (R.K.); (S.F.)
| | - Sandra Freitag
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (V.G.); (L.C.); (R.K.); (S.F.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (V.G.); (L.C.); (R.K.); (S.F.)
| |
Collapse
|
3
|
Tomalty D, Giovannetti O, Velikonja L, Munday J, Kaufmann M, Iaboni N, Jamzad A, Rubino R, Fichtinger G, Mousavi P, Nicol CJB, Rudan JF, Adams MA. Molecular characterization of human peripheral nerves using desorption electrospray ionization mass spectrometry imaging. J Anat 2023; 243:758-769. [PMID: 37264225 PMCID: PMC10557387 DOI: 10.1111/joa.13909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/11/2023] [Accepted: 05/20/2023] [Indexed: 06/03/2023] Open
Abstract
Desorption electrospray ionization mass spectrometry imaging (DESI-MSI) is a molecular imaging method that can be used to elucidate the small-molecule composition of tissues and map their spatial information using two-dimensional ion images. This technique has been used to investigate the molecular profiles of variety of tissues, including within the central nervous system, specifically the brain and spinal cord. To our knowledge, this technique has yet to be applied to tissues of the peripheral nervous system (PNS). Data generated from such analyses are expected to advance the characterization of these structures. The study aimed to: (i) establish whether DESI-MSI can discriminate the molecular characteristics of peripheral nerves and distinguish them from surrounding tissues and (ii) assess whether different peripheral nerve subtypes are characterized by unique molecular profiles. Four different nerves for which are known to carry various nerve fiber types were harvested from a fresh cadaveric donor: mixed, motor and sensory (sciatic and femoral); cutaneous, sensory (sural); and autonomic (vagus). Tissue samples were harvested to include the nerve bundles in addition to surrounding connective tissue. Samples were flash-frozen, embedded in optimal cutting temperature compound in cross-section, and sectioned at 14 μm. Following DESI-MSI analysis, identical tissue sections were stained with hematoxylin and eosin. In this proof-of-concept study, a combination of multivariate and univariate statistical methods was used to evaluate molecular differences between the nerve and adjacent tissue and between nerve subtypes. The acquired mass spectral profiles of the peripheral nerve samples presented trends in ion abundances that seemed to be characteristic of nerve tissue and spatially corresponded to the associated histology of the tissue sections. Principal component analysis (PCA) supported the separation of the samples into distinct nerve and adjacent tissue classes. This classification was further supported by the K-means clustering analysis, which showed separation of the nerve and background ions. Differences in ion expression were confirmed using ANOVA which identified statistically significant differences in ion expression between the nerve subtypes. The PCA plot suggested some separation of the nerve subtypes into four classes which corresponded with the nerve types. This was supported by the K-means clustering. Some overlap in classes was noted in these two clustering analyses. This study provides emerging evidence that DESI-MSI is an effective tool for metabolomic profiling of peripheral nerves. Our results suggest that peripheral nerves have molecular profiles that are distinct from the surrounding connective tissues and that DESI-MSI may be able to discriminate between nerve subtypes. DESI-MSI of peripheral nerves may be a valuable technique that could be used to improve our understanding of peripheral nerve anatomy and physiology. The ability to utilize ambient mass spectrometry techniques in real time could also provide an unprecedented advantage for surgical decision making, including in nerve-sparing procedures in the future.
Collapse
Affiliation(s)
- Diane Tomalty
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Olivia Giovannetti
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Leah Velikonja
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Jasica Munday
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Martin Kaufmann
- Department of SurgeryQueen's UniversityKingstonOntarioCanada
- Gastrointestinal Diseases Research UnitKingston Health Sciences CenterKingstonOntarioCanada
| | - Natasha Iaboni
- Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - Amoon Jamzad
- School of ComputingQueen's UniversityKingstonOntarioCanada
| | - Rachel Rubino
- Division of Cancer Biology and GeneticsQueen's Cancer Research InstituteKingstonOntarioCanada
| | | | - Parvin Mousavi
- School of ComputingQueen's UniversityKingstonOntarioCanada
| | - Christopher J. B. Nicol
- Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
- Division of Cancer Biology and GeneticsQueen's Cancer Research InstituteKingstonOntarioCanada
| | - John F. Rudan
- Department of SurgeryQueen's UniversityKingstonOntarioCanada
| | - Michael A. Adams
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
4
|
Tschang M, Kumar S, Young W, Schachner M, Theis T. Small Organic Compounds Mimicking the Effector Domain of Myristoylated Alanine-Rich C-Kinase Substrate Stimulate Female-Specific Neurite Outgrowth. Int J Mol Sci 2023; 24:14271. [PMID: 37762575 PMCID: PMC10532424 DOI: 10.3390/ijms241814271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Myristoylated alanine-rich C-kinase substrate (MARCKS) is a critical member of a signaling cascade that influences disease-relevant neural functions such as neural growth and plasticity. The effector domain (ED) of MARCKS interacts with the extracellular glycan polysialic acid (PSA) through the cell membrane to stimulate neurite outgrowth in cell culture. We have shown that a synthetic ED peptide improves functional recovery after spinal cord injury in female but not male mice. However, peptides themselves are unstable in therapeutic applications, so we investigated more pharmacologically relevant small organic compounds that mimic the ED peptide to maximize therapeutic potential. Using competition ELISAs, we screened small organic compound libraries to identify molecules that structurally and functionally mimic the ED peptide of MARCKS. Since we had shown sex-specific effects of MARCKS on spinal cord injury recovery, we assayed neuronal viability as well as neurite outgrowth from cultured cerebellar granule cells of female and male mice separately. We found that epigallocatechin, amiodarone, sertraline, tegaserod, and nonyloxytryptamine bind to a monoclonal antibody against the ED peptide, and compounds stimulate neurite outgrowth in cultured cerebellar granule cells of female mice only. Therefore, a search for compounds that act in males appears warranted.
Collapse
Affiliation(s)
- Monica Tschang
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08844, USA;
| | - Wise Young
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| |
Collapse
|
5
|
Congiu L, Granato V, Jakovcevski I, Kleene R, Fernandes L, Freitag S, Kneussel M, Schachner M, Loers G. Mice Mutated in the Third Fibronectin Domain of L1 Show Enhanced Hippocampal Neuronal Cell Death, Astrogliosis and Alterations in Behavior. Biomolecules 2023; 13:776. [PMID: 37238646 PMCID: PMC10216033 DOI: 10.3390/biom13050776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Adhesion molecules play major roles in cell proliferation, migration, survival, neurite outgrowth and synapse formation during nervous system development and in adulthood. The neural cell adhesion molecule L1 contributes to these functions during development and in synapse formation and synaptic plasticity after trauma in adulthood. Mutations of L1 in humans result in L1 syndrome, which is associated with mild-to-severe brain malformations and mental disabilities. Furthermore, mutations in the extracellular domain were shown to cause a severe phenotype more often than mutations in the intracellular domain. To explore the outcome of a mutation in the extracellular domain, we generated mice with disruption of the dibasic sequences RK and KR that localize to position 858RKHSKR863 in the third fibronectin type III domain of murine L1. These mice exhibit alterations in exploratory behavior and enhanced marble burying activity. Mutant mice display higher numbers of caspase 3-positive neurons, a reduced number of principle neurons in the hippocampus, and an enhanced number of glial cells. Experiments suggest that disruption of the dibasic sequence in L1 results in subtle impairments in brain structure and functions leading to obsessive-like behavior in males and reduced anxiety in females.
Collapse
Affiliation(s)
- Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Viviana Granato
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Sandra Freitag
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Matthias Kneussel
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| |
Collapse
|
6
|
Ubiquitous Neural Cell Adhesion Molecule (NCAM): Potential Mechanism and Valorisation in Cancer Pathophysiology, Drug Targeting and Molecular Transductions. Mol Neurobiol 2022; 59:5902-5924. [PMID: 35831555 DOI: 10.1007/s12035-022-02954-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/02/2022] [Indexed: 10/17/2022]
Abstract
Neural cell adhesion molecule, an integrated molecule of immunoglobulin protein superfamily involved in cell-cell adhesion, undergoes various structural modifications through numerous temporal-spatial regulations that generously alter their expressions on cell surfaces. These varied expression patterns are mostly envisioned in the morphogenesis and innervations of different human organs and systems. The considerable role of NCAM in neurite growth, brain development and etc. and its altered expression of NCAM in proliferating tumour cells and metastasis of various human melanomas clearly substantiate its appropriateness as a cell surface marker for diagnosis and potential target for several therapeutic moieties. This characteristic behaviour of NCAM is confined to its novel biochemistry, structural properties, signalling interactions and polysialylation. In particular, the characteristic expressions of NCAM are mainly attributed by its polysialylation, a post-translational modification that attaches polysialyl groups to the NCAM. The altered expression of NCAM on cell surface develops curiosity amidst pharmaceutical scientists, which drives them to understand its role of such expressions in various human melanomas and to elucidate the promising therapeutic strategies that are currently available to target NCAM appositely. Therefore, this review article is articulated with an insight on the altered expressions of NCAM, the clinical significances and the consequences of such atypical expression patterns in various human organs and systems.
Collapse
|
7
|
Wang B, Li Z, Li J, Shao Q, Qin L. Sialin mediates submandibular gland regeneration ability by affecting polysialic acid synthesis. Oral Dis 2022. [PMID: 35593110 DOI: 10.1111/odi.14256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/27/2022] [Accepted: 05/12/2022] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Sialin is a multifunctional molecule with a well-described role in physiological equilibrium regulation. The aim of this study was to elucidate the role of sialin in salivary glands regeneration. MATERIALS AND METHODS Submandibular gland duct ligation/deligation of rat was performed to develop a rat model of submandibular gland regeneration. Phenotype changes were investigated using western blotting and quantitative real-time polymerase chain reaction, as well as immunohistochemical staining. LV-slc17a5-RNAi vectors were injected into the submandibular glands via retroductal instillation to establish a stable sialin knockdown model. RESULTS Submandibular gland tissue structure could completely restore 28 days after duct deligation, when the duct had been ligated for 7 days. The expression of sialin, polysialic acid, and polysialyltransferase IV was significantly increased on day 0 after duct deligation, and it returned to the level of the control group at day 28. Moreover, sialin knockdown could weakened gland regeneration by reducing polysialic acid synthesis. Supplementing drinking water with polysialic acid precursors (ManNAc) in drinking water could partially rescue submandibular gland regeneration in sialin knockdown rats. CONCLUSION These data indicated that sialin was vital for submandibular gland regeneration which mediated the process of gland regeneration by affecting the polysialic acid synthesis.
Collapse
Affiliation(s)
- Bin Wang
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Zhilin Li
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China.,Department of Head and Neck Oncology, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Qi Shao
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China.,Department of Oral and Maxillofacial Surgery, Changsha Stomatological Hospital, You Yi Road No.389, Changsha, China
| | - Lizheng Qin
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Congiu L, Granato V, Loers G, Kleene R, Schachner M. Mitochondrial and Neuronal Dysfunctions in L1 Mutant Mice. Int J Mol Sci 2022; 23:ijms23084337. [PMID: 35457156 PMCID: PMC9026747 DOI: 10.3390/ijms23084337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 12/04/2022] Open
Abstract
Adhesion molecules regulate cell proliferation, migration, survival, neuritogenesis, synapse formation and synaptic plasticity during the nervous system’s development and in the adult. Among such molecules, the neural cell adhesion molecule L1 contributes to these functions during development, and in synapse formation, synaptic plasticity and regeneration after trauma. Proteolytic cleavage of L1 by different proteases is essential for these functions. A proteolytic fragment of 70 kDa (abbreviated L1-70) comprising part of the extracellular domain and the transmembrane and intracellular domains was shown to interact with mitochondrial proteins and is suggested to be involved in mitochondrial functions. To further determine the role of L1-70 in mitochondria, we generated two lines of gene-edited mice expressing full-length L1, but no or only low levels of L1-70. We showed that in the absence of L1-70, mitochondria in cultured cerebellar neurons move more retrogradely and exhibit reduced mitochondrial membrane potential, impaired Complex I activity and lower ATP levels compared to wild-type littermates. Neither neuronal migration, neuronal survival nor neuritogenesis in these mutants were stimulated with a function-triggering L1 antibody or with small agonistic L1 mimetics. These results suggest that L1-70 is important for mitochondrial homeostasis and that its absence contributes to the L1 syndrome phenotypes.
Collapse
Affiliation(s)
- Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (L.C.); (V.G.); (G.L.); (R.K.)
| | - Viviana Granato
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (L.C.); (V.G.); (G.L.); (R.K.)
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (L.C.); (V.G.); (G.L.); (R.K.)
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; (L.C.); (V.G.); (G.L.); (R.K.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
- Correspondence: ; Tel.: +1-848-445-1780
| |
Collapse
|
9
|
Jakovcevski I, von Düring M, Lutz D, Vulović M, Hamad M, Reiss G, Förster E, Schachner M. Mice lacking perforin have improved regeneration of the injured femoral nerve. Neural Regen Res 2022; 17:1802-1808. [PMID: 35017441 PMCID: PMC8820721 DOI: 10.4103/1673-5374.332152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The role that the immune system plays after injury of the peripheral nervous system is still not completely understood. Perforin, a natural killer cell- and T-lymphocyte-derived enzyme that mediates cytotoxicity, plays important roles in autoimmune diseases, infections and central nervous system trauma, such as spinal cord injury. To dissect the roles of this single component of the immune response to injury, we tested regeneration after femoral nerve injury in perforin-deficient (Pfp–/–) and wild-type control mice. Single frame motion analysis showed better motor recovery in Pfp–/– mice compared with control mice at 4 and 8 weeks after injury. Retrograde tracing of the motoneuron axons regrown into the motor nerve branch demonstrated more correctly projecting motoneurons in the spinal cord of Pfp–/– mice compared with wild-types. Myelination of regrown axons measured by g-ratio was more extensive in Pfp–/– than in wild-type mice in the motor branch of the femoral nerve. Pfp–/– mice displayed more cholinergic synaptic terminals around cell bodies of spinal motoneurons after injury than the injured wild-types. We histologically analyzed lymphocyte infiltration 10 days after surgery and found that in Pfp–/– mice the number of lymphocytes in the regenerating nerves was lower than in wild-types, suggesting a closed blood-nerve barrier in Pfp–/– mice. We conclude that perforin restricts motor recovery after femoral nerve injury owing to decreased survival of motoneurons and reduced myelination.
Collapse
Affiliation(s)
- Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, Witten, Germany
| | - Monika von Düring
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - David Lutz
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - Maja Vulović
- Department of Anatomy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Mohammad Hamad
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, Witten, Germany
| | - Gebhard Reiss
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, Witten, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
10
|
Parker BJ, Rhodes DI, O'Brien CM, Rodda AE, Cameron NR. Nerve guidance conduit development for primary treatment of peripheral nerve transection injuries: A commercial perspective. Acta Biomater 2021; 135:64-86. [PMID: 34492374 DOI: 10.1016/j.actbio.2021.08.052] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
Commercial nerve guidance conduits (NGCs) for repair of peripheral nerve discontinuities are of little use in gaps larger than 30 mm, and for smaller gaps they often fail to compete with the autografts that they are designed to replace. While recent research to develop new technologies for use in NGCs has produced many advanced designs with seemingly positive functional outcomes in animal models, these advances have not been translated into viable clinical products. While there have been many detailed reviews of the technologies available for creating NGCs, none of these have focussed on the requirements of the commercialisation process which are vital to ensure the translation of a technology from bench to clinic. Consideration of the factors essential for commercial viability, including regulatory clearance, reimbursement processes, manufacturability and scale up, and quality management early in the design process is vital in giving new technologies the best chance at achieving real-world impact. Here we have attempted to summarise the major components to consider during the development of emerging NGC technologies as a guide for those looking to develop new technology in this domain. We also examine a selection of the latest academic developments from the viewpoint of clinical translation, and discuss areas where we believe further work would be most likely to bring new NGC technologies to the clinic. STATEMENT OF SIGNIFICANCE: NGCs for peripheral nerve repairs represent an adaptable foundation with potential to incorporate modifications to improve nerve regeneration outcomes. In this review we outline the regulatory processes that functionally distinct NGCs may need to address and explore new modifications and the complications that may need to be addressed during the translation process from bench to clinic.
Collapse
Affiliation(s)
- Bradyn J Parker
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Research Way, Clayton, Victoria 3168, Australia
| | - David I Rhodes
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia; ReNerve Pty. Ltd., Brunswick East 3057, Australia
| | - Carmel M O'Brien
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Research Way, Clayton, Victoria 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and innovation Precinct (STRIP), Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Andrew E Rodda
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia
| | - Neil R Cameron
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, Victoria 3800, Australia; School of Engineering, University of Warwick, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
11
|
Lünemann JD, von Gunten S, Neumann H. Targeting sialylation to treat central nervous system diseases. Trends Pharmacol Sci 2021; 42:998-1008. [PMID: 34607695 DOI: 10.1016/j.tips.2021.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 02/03/2023]
Abstract
Sialic acid-binding immunoglobulin-type lectins (SIGLECs) are membrane receptors that are preferentially expressed on immune cells and recognize sialylated proteins, lipids, and RNA. Sialic acids and signaling through SIGLECs are increasingly recognized for their essential roles in immune system homeostasis as well as nervous system development, plasticity, and repair. Dysregulated sialylation and SIGLEC dysfunctions contribute to several chronic diseases of the central nervous system (CNS) in which current therapeutic options are very limited. While only a few therapies targeting SIGLECs are currently being tested in clinical trials, the area emerged as one of the most dynamic and active fields in glycobiology and drug development. This review highlights recent insights into sialic acid and SIGLEC function in CNS pathologies and illustrates opportunities and challenges for the development of sialic acid-based and SIGLEC-targeted therapies for neurological diseases.
Collapse
Affiliation(s)
- Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| | | | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Pearse DD, Rao SNR, Morales AA, Wakarchuk W, Rutishauser U, El-Maarouf A, Ghosh M. Engineering polysialic acid on Schwann cells using polysialyltransferase gene transfer or purified enzyme exposure for spinal cord injury transplantation. Neurosci Lett 2021; 748:135690. [PMID: 33540059 DOI: 10.1016/j.neulet.2021.135690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/23/2020] [Accepted: 01/25/2021] [Indexed: 11/29/2022]
Abstract
Polysialic acid (PolySia) is a critical post-translational modification on the neural cell adhesion molecule (NCAM, a.k.a., CD56), important for cell migration and axon growth during nervous system development, plasticity and repair. PolySia induction on Schwann cells (SCs) enhances their migration, axon growth support and ability to improve functional recovery after spinal cord injury (SCI) transplantation. In the current investigation two methods of PolySia induction on SCs, lentiviral vector transduction of the mouse polysialytransferase gene ST8SIA4 (LV-PST) or enzymatic engineering with a recombinant bacterial PST (PSTNm), were examined comparatively for their effects on PolySia induction, SC migration, the innate immune response and axon growth after acute SCI. PSTNm produced significant PolySia induction and a greater diversity of surface molecule polysialylation on SCs as evidenced by immunoblot. In the scratch wound assay, PSTNm was superior to LV-PST in the promotion of SC migration and gap closure. At 24 h after SCI transplantation, PolySia induction on SCs was most pronounced with LV-PST. Co-delivery of PSTNm with SCs, but not transient cell exposure, led to broader induction of PolySia within the injured spinal cord due to polysialylation upon both host cells and transplanted SCs. The innate immune response after SCI, measured by CD68 immunoreactivity, was similar among PolySia induction methods. LV-PST or PSTNm co-delivery with SCs provided a similar enhancement of SC migration and axon growth support above that of unmodified SCs. These studies demonstrate that LV-PST and PSTNm provide comparable acute effects on SC polysialation, the immune response and neurorepair after SCI.
Collapse
Affiliation(s)
- Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL, 33136, USA.
| | - Sudheendra N R Rao
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejo A Morales
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Warren Wakarchuk
- Department of Biological Sciences, University of Alberta, Edmonton, AB, TG6 2E9, Canada
| | - Urs Rutishauser
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL, 33136, USA.
| |
Collapse
|
13
|
Sytnyk V, Leshchyns'ka I, Schachner M. Neural glycomics: the sweet side of nervous system functions. Cell Mol Life Sci 2021; 78:93-116. [PMID: 32613283 PMCID: PMC11071817 DOI: 10.1007/s00018-020-03578-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/06/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
The success of investigations on the structure and function of the genome (genomics) has been paralleled by an equally awesome progress in the analysis of protein structure and function (proteomics). We propose that the investigation of carbohydrate structures that go beyond a cell's metabolism is a rapidly developing frontier in our expanding knowledge on the structure and function of carbohydrates (glycomics). No other functional system appears to be suited as well as the nervous system to study the functions of glycans, which had been originally characterized outside the nervous system. In this review, we describe the multiple studies on the functions of LewisX, the human natural killer cell antigen-1 (HNK-1), as well as oligomannosidic and sialic (neuraminic) acids. We attempt to show the sophistication of these structures in ontogenetic development, synaptic function and plasticity, and recovery from trauma, with a view on neurodegeneration and possibilities to ameliorate deterioration. In view of clinical applications, we emphasize the need for glycomimetic small organic compounds which surpass the usefulness of natural glycans in that they are metabolically more stable, more parsimonious to synthesize or isolate, and more advantageous for therapy, since many of them pass the blood brain barrier and are drug-approved for treatments other than those in the nervous system, thus allowing a more ready access for application in neurological diseases. We describe the isolation of such mimetic compounds using not only Western NIH, but also traditional Chinese medical libraries. With this review, we hope to deepen the interests in this exciting field.
Collapse
Affiliation(s)
- Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia.
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, 515041, Guangdong, China
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
14
|
Kalotra S, Kaur G. PSA mimetic 5-nonyloxytryptamine protects cerebellar neurons against glutamate induced excitotoxicity: An in vitro perspective. Neurotoxicology 2020; 82:69-81. [PMID: 33197482 DOI: 10.1016/j.neuro.2020.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 12/29/2022]
Abstract
PSA-NCAM is a molecule of therapeutic interest for its key role in promoting neuritogenesis and synaptic plasticity. The current study was aimed to investigate the neuroregenerative potential of 5-nonyloxytryptamine (5-NOT) as PSA mimetic compound against glutamate induced excitotoxicity. 2D and 3D cultures of cerebellar neurons challenged with glutamate were used to ascertain the effect of 5-NOT on neurite outgrowth, migration and expression of neuronal plasticity markers. Glutamate excitotoxicity is one of the major underlying pathological factor responsible for neurodegeneration in various neurological disorders such as trauma, stroke, ischemia, epilepsy and neurodegenerative diseases.5-NOT treatment was observed to promote axonal growth and defasiculation in glutamate challenged neurons as well as promoted the migration of cerebellar neurons in both wound scratched area and cerebellar explant cultures. Further, 5-NOT treatment upregulated the expression of synaptic plasticity and cell survival pathway proteins which showed reduced expression after glutamate induced excitotoxicity. Thus, this preliminary data reveals thatPSA-mimetic,5-NOT may prove to be a potential neuroprotective candidate for neurodegenerative diseases.
Collapse
Affiliation(s)
- Shikha Kalotra
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| |
Collapse
|
15
|
Saini V, Kaur T, Kalotra S, Kaur G. The neuroplasticity marker PSA-NCAM: Insights into new therapeutic avenues for promoting neuroregeneration. Pharmacol Res 2020; 160:105186. [DOI: 10.1016/j.phrs.2020.105186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
|
16
|
Schwann Cell Role in Selectivity of Nerve Regeneration. Cells 2020; 9:cells9092131. [PMID: 32962230 PMCID: PMC7563640 DOI: 10.3390/cells9092131] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries result in the loss of the motor, sensory and autonomic functions of the denervated segments of the body. Neurons can regenerate after peripheral axotomy, but inaccuracy in reinnervation causes a permanent loss of function that impairs complete recovery. Thus, understanding how regenerating axons respond to their environment and direct their growth is essential to improve the functional outcome of patients with nerve lesions. Schwann cells (SCs) play a crucial role in the regeneration process, but little is known about their contribution to specific reinnervation. Here, we review the mechanisms by which SCs can differentially influence the regeneration of motor and sensory axons. Mature SCs express modality-specific phenotypes that have been associated with the promotion of selective regeneration. These include molecular markers, such as L2/HNK-1 carbohydrate, which is differentially expressed in motor and sensory SCs, or the neurotrophic profile after denervation, which differs remarkably between SC modalities. Other important factors include several molecules implicated in axon-SC interaction. This cell–cell communication through adhesion (e.g., polysialic acid) and inhibitory molecules (e.g., MAG) contributes to guiding growing axons to their targets. As many of these factors can be modulated, further research will allow the design of new strategies to improve functional recovery after peripheral nerve injuries.
Collapse
|
17
|
Theis T, Kumar S, Wei E, Nguyen J, Glynos V, Paranjape N, Askarifirouzjaei H, Khajouienejad L, Berthiaume F, Young W, Schachner M. Myristoylated alanine-rich C-kinase substrate effector domain peptide improves sex-specific recovery and axonal regrowth after spinal cord injury. FASEB J 2020; 34:12677-12690. [PMID: 32729988 DOI: 10.1096/fj.202000026rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 11/11/2022]
Abstract
Myristoylated alanine-rich C-kinase substrate (MARCKS) is an intracellular receptor for polysialic acid. MARCKS supports development, synaptic plasticity, and regeneration after injury. MARCKS binds with its functionally essential effector domain (ED) to polysialic acid. A 25-mer peptide comprising the ED of MARCKS stimulates neuritogenesis of primary hippocampal neurons after addition to the culture. This motivated us to investigate whether ED peptide has similar effects in spinal cord injury. ED peptide supported recovery and regrowth of monoaminergic axons in female, but not in male mice. Sex-specific differences in response to ED peptide application also occurred in cultured neurons. In female but not male neurons, the ED peptide enhanced neurite outgrowth that could be suppressed by inhibitors of the estrogen receptors α and β, fibroblast growth factor receptor-1, protein kinase C, and matrix metalloproteinase 2. In addition, we observed female-specific elevation of phosphorylated MARCKS levels after ED peptide treatment. In male neurons, the ED peptide enhanced neuritogenesis in the presence of an androgen receptor inhibitor to the extent seen in ED peptide-treated female neurons. However, inhibition of androgen receptor did not lead to increased phosphorylation of MARCKS. These results provide insights into the functions of a novel compound contributing to gender-dependent regeneration.
Collapse
Affiliation(s)
- Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Elena Wei
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Jennifer Nguyen
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Vicci Glynos
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Nikita Paranjape
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Hadi Askarifirouzjaei
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Leila Khajouienejad
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Wise Young
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
18
|
Chen SX, He JH, Mi YJ, Shen HF, Schachner M, Zhao WJ. A mimetic peptide of α2,6-sialyllactose promotes neuritogenesis. Neural Regen Res 2020; 15:1058-1065. [PMID: 31823885 PMCID: PMC7034278 DOI: 10.4103/1673-5374.270313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 06/21/2019] [Accepted: 07/28/2019] [Indexed: 02/05/2023] Open
Abstract
Oxidative stress contributes to the pathogenesis of neurodegenerative diseases. With the aim to find reagents that reduce oxidative stress, a phage display library was screened for peptides mimicking α2,6-sialyllactose (6'-SL), which is known to beneficially influence neural functions. Using Sambucus nigra lectin, which specifically binds to 6'-SL, we screened a phage display library and found a peptide comprising identical sequences of 12 amino acids. Mimetic peptide, reverse peptide and scrambled peptide were tested for inhibition of 6'-SL binding to the lectin. Indeed, lectin binding to 6'-SL was inhibited by the most frequently identified mimetic peptide, but not by the reverse or scrambled peptides, showing that this peptide mimics 6'-SL. Functionally, mimetic peptide, but not the reverse or scrambled peptides, increased viability and expression of neural cell adhesion molecule L1 in SK-N-SH human neuroblastoma cells, and promoted survival and neurite outgrowth of cultured mouse cerebellar granule neurons challenged by H2O2-induced oxidative stress. The combined results indicate that the 6'-SL mimetic peptide promotes neuronal survival and neuritogenesis, thus raising hopes for the treatment of neurodegenerative diseases. This study was approved by the Medical Ethics Committee of Shantou University Medical College, China (approval No. SUMC 2014-004) on February 20, 2014.
Collapse
Affiliation(s)
- Shuang-Xi Chen
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Jia-Hui He
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Yong-Jian Mi
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Neurology, Chongqing Qijiang Renmin Hospital, Chongqing, China
| | - Hui-Fan Shen
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Correspondence to: Melitta Schachner, ; Wei-Jiang Zhao,
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, China
- Correspondence to: Melitta Schachner, ; Wei-Jiang Zhao,
| |
Collapse
|
19
|
Kalotra S, Saini V, Singh H, Sharma A, Kaur G. 5-Nonyloxytryptamine oxalate-embedded collagen-laminin scaffolds augment functional recovery after spinal cord injury in mice. Ann N Y Acad Sci 2019; 1465:99-116. [PMID: 31800108 DOI: 10.1111/nyas.14279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/03/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022]
Abstract
Polysialic acid (PSA) is crucial for the induction and maintenance of nervous system plasticity and repair after injury. In order to exploit the immense therapeutic potential of PSA, previous studies have focused on the identification and development of peptide-based or synthetic PSA mimetics. 5-Nonyloxytryptamine (5-NOT) has been previously reported as a PSA-mimicking compound for promoting functional recovery after spinal cord injury in mice. In order to explore the neuroregeneration potential of 5-NOT, the current study was based on a biomaterial approach using collagen-laminin (C/L) scaffolds. In in vitro studies, 5-NOT was observed to promote neurite outgrowth, migration, and fasciculation in cerebellar neuronal cells, whereas in 3D cell cultures it showed more ramification and complex Sholl profiles. 5-NOT promoted the survival and neurite length of cortical neurons when cocultured with glutamate-challenged astrocytes. In in vivo studies, spinal cord compression injury mice were used with immediate application of C/L hydrogels impregnated with 5-NOT. C/L + 5-NOT-treated mice demonstrated ∼75% of motor recovery 14 days after injury. Furthermore, this effect was shown to be dependent on the ERK-MAPK pathway and augmentation of cell survival. Thus, based on a biomaterial approach, our current study provides new insight for 5-NOT-containing hydrogels as a promising candidate to speed up recovery after central nervous system injuries.
Collapse
Affiliation(s)
- Shikha Kalotra
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vedangana Saini
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Harpal Singh
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Anuradha Sharma
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
20
|
Raslan A, Salem MAM, Al‐Hussaini A, Guntinas‐Lichius O, Irintchev A. Brief Electrical Stimulation Improves Functional Recovery After Femoral But Not After Facial Nerve Injury in Rats. Anat Rec (Hoboken) 2019; 302:1304-1313. [DOI: 10.1002/ar.24127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/08/2018] [Accepted: 09/11/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Ashraf Raslan
- Department of OtorhinolaryngologyJena University Hospital Jena Germany
- Department of OtorhinolaryngologyAssiut University Assiut Egypt
| | | | | | | | - Andrey Irintchev
- Department of OtorhinolaryngologyJena University Hospital Jena Germany
| |
Collapse
|
21
|
Wang XJ, Peng CH, Zhang S, Xu XL, Shu GF, Qi J, Zhu YF, Xu DM, Kang XQ, Lu KJ, Jin FY, Yu RS, Ying XY, You J, Du YZ, Ji JS. Polysialic-Acid-Based Micelles Promote Neural Regeneration in Spinal Cord Injury Therapy. NANO LETTERS 2019; 19:829-838. [PMID: 30605619 DOI: 10.1021/acs.nanolett.8b04020] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Spinal cord injury (SCI) routinely causes the immediate loss and disruption of neurons followed by complicated secondary injuries, including inflammation, oxidative stress, and dense glial scar formation. Inhibitory factors in the lesion scar and poor intrinsic neural regeneration capacity restrict functional recovery after injury. Minocycline, which has neuroprotective activity, can alleviate secondary injury, but the long-term administration of this drug may cause toxicity. Polysialic acid (PSA) is a large cell-surface carbohydrate that is critical for central nervous system development and is capable of promoting precursor cell migration, axon path finding, and synaptic remodeling; thus, PSA plays a vital role in tissue repair and regeneration. Here, we developed a PSA-based minocycline-loaded nanodrug delivery system (PSM) for the synergistic therapy of spinal cord injury. The prepared PSM exerted marked anti-inflammatory and neuroprotective activities both in vitro and in vivo. The administration of PSM could significantly protect neurons and myelin sheaths from damage, reduce the formation of glial scar, recruit endogenous neural stem cells to the lesion site, and promote the regeneration of neurons and the extension of long axons throughout the glial scar, thereby largely improving the locomotor function of SCI rats and exerting a superior therapeutic effect. The findings might provide a novel strategy for SCI synergistic therapy and the utilization of PSA in other central nervous system diseases.
Collapse
Affiliation(s)
- Xiao-Juan Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Chen-Han Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Shuo Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Gao-Feng Shu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research , Lishui Hospital of Zhejiang University , Lishui 323000 , China
| | - Jing Qi
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Ya-Fang Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - De-Min Xu
- Department of Radiology, Second Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310009 , PR China
| | - Xu-Qi Kang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Kong-Jun Lu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Fei-Yang Jin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310009 , PR China
| | - Xiao-Ying Ying
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Jian You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , PR China
| | - Jian-Song Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research , Lishui Hospital of Zhejiang University , Lishui 323000 , China
| |
Collapse
|
22
|
Broguiere N, Husch A, Palazzolo G, Bradke F, Madduri S, Zenobi-Wong M. Macroporous hydrogels derived from aqueous dynamic phase separation. Biomaterials 2019; 200:56-65. [PMID: 30772759 DOI: 10.1016/j.biomaterials.2019.01.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
Abstract
A method to generate injectable macroporous hydrogels based on partitioning of polyethylene glycol (PEG) and high viscous polysaccharides is presented. Step growth polymerization of PEG was used to initiate a phase separation and the formation of a connected macroporous network with tunable dimensions. The possibilities and physical properties of this new category of materials were examined, and then applied to address some challenges in neural engineering. First, non-degradable macroporous gels were shown to support rapid neurite extension from encapsulated dorsal root ganglia (DRGs) with unprecedented long-term stability. Then, dissociated primary rat cortical neurons could be encapsulated with >95% viability, and extended neurites at the fast rate of ≈100 μm/day and formed synapses, resulting in functional, highly viable and long-term stable 3D neural networks in the synthetic extracellular matrix (ECM). Adhesion cues were found unnecessary provided the gels have optimal physical properties. Normal electrophysiological properties were confirmed on 3D cultured mouse hippocampal neurons. Finally, the macroporous gels supported axonal growth in a rat sciatic nerve injury model when used as a conduit filling. The combination of injectability, tunable pore size, stability, connectivity, transparency, cytocompatibility and biocompatibility, makes this new class of materials attractive for a wide range of applications.
Collapse
Affiliation(s)
- Nicolas Broguiere
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Andreas Husch
- Axon Growth and Regeneration Laboratory, German Center for Neurodegenerative Diseases, Sigmund-Freud-Str. 27, 53127, Bonn, Germany
| | - Gemma Palazzolo
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Frank Bradke
- Axon Growth and Regeneration Laboratory, German Center for Neurodegenerative Diseases, Sigmund-Freud-Str. 27, 53127, Bonn, Germany
| | - Srinivas Madduri
- Center for Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, Gewerbestrasse-14, 4123 Allschwil Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Klinikum 1, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland.
| |
Collapse
|
23
|
Kleene R, Loers G, Jakovcevski I, Mishra B, Schachner M. Histone H1 improves regeneration after mouse spinal cord injury and changes shape and gene expression of cultured astrocytes. Restor Neurol Neurosci 2019; 37:291-313. [PMID: 31227672 DOI: 10.3233/rnn-190903] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We have shown that histone H1 is a binding partner for polysialic acid (PSA) and that it improves functional recovery, axon regrowth/sprouting, and target reinnervation after mouse femoral nerve injury. OBJECTIVE Here, we analyzed whether histone H1 affects functional recovery, axon regrowth/sprouting, and target reinnervation after spinal cord injury of adult mice. Furthermore, we tested in vitro histone H1's effect on astrocytic gene expression, cell shape and migration as well as on cell survival of cultured motoneurons. METHODS We applied histone H1 to compressed spinal cord and determined functional recovery and number of fibrillary acidic protein (GFAP)- and neuron-glial antigen 2 (NG2)- positive glial cells, which contribute to glial scarring. Histone H1's effect on migration of astrocytes, astrocytic gene expression and motoneuronal survival was determined using scratch-wounded astroglial monolayer cultures, astrocyte cultures for microarray analysis, and motoneuron cell culture under oxidative stress conditions, respectively. RESULTS Histone H1 application improves locomotor functions and enhances monoaminergic and cholinergic reinnervation of the spinal cord. Expression levels of GFAP and NG2 around the lesion site were decreased in histone H1-treated mice relative to vehicle-treated mice six weeks after injury. Histone H1 reduced astrocytic migration, changed the shape of GFAP- and NG2-positive glial cells and altered gene expression. Gene ontology enrichment analysis indicated that in particular genes coding for proteins involved in proliferation, differentiation, migration and apoptosis are dysregulated. The up- and down-regulation of distinct genes was confirmed by qPCR and Western blot analysis. Moreover, histone H1 reduced hydrogen peroxide-induced cell death of cultured motoneurons. CONCLUSIONS The combined observations indicate that histone H1 locally applied to the lesion site, improves regeneration after spinal cord injury. Some of these beneficial functions of histone H1 in vivo and in vitro can be attributed to its interaction with PSA-carrying neural cell adhesion molecule.
Collapse
Affiliation(s)
- Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Igor Jakovcevski
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
24
|
Zhang S, Wang XJ, Li WS, Xu XL, Hu JB, Kang XQ, Qi J, Ying XY, You J, Du YZ. Polycaprolactone/polysialic acid hybrid, multifunctional nanofiber scaffolds for treatment of spinal cord injury. Acta Biomater 2018; 77:15-27. [PMID: 30126591 DOI: 10.1016/j.actbio.2018.06.038] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 12/11/2022]
Abstract
Scaffold-based tissue engineering is widely used for spinal cord injury (SCI) treatment by creating supporting and guiding neuronal tissue regeneration. However, how to enhance the axonal regeneration capacity following SCI still remains a challenge. Polysialic acid (PSA), a natural, biodegradable polysaccharide, has been increasingly explored for controlling central nervous system (CNS) development by regulating cell adhesive properties and promoting axonal growth. Here, a polycaprolactone (PCL)/PSA hybrid nanofiber scaffold encapsulating glucocorticoid methylprednisolone (MP) is developed for SCI treatment. Rat models with spinal cord transection is established and the PCL/PSA/MP scaffold is transplanted into lesion area. PCL/PSA/MP scaffold decreases tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) release by inhibiting ionized calcium-binding adapter molecule 1 (Iba1) positive microglia/macrophage activation and reduces apoptosis-associated Caspase-3 protein expression. In addition, the PCL/PSA/MP scaffold inhibits axonal demyelination and glial fibrillary acidic protein (GFAP) expression, increases neurofilament 200 (NF-200) expression and improves functional outcome by Basso, Beattie and Bresnahan (BBB) test. These results demonstrate the therapeutic potential of PSA hybrid nanofiber scaffold in promoting axonal growth and enhancing the functional recovery following SCI. STATEMENT OF SIGNIFICANCE Scaffold-based tissue engineering is widely used for spinal cord injury (SCI) treatment by creating supporting and guiding neuronal tissue regeneration. And how to enhance the axonal regeneration capacity following SCI still remains a challenge. Polysialic acid (PSA), a natural, biodegradable polysaccharide, has been increasingly explored for controlling central nervous system (CNS) development by regulating cell adhesive properties and promoting axonal growth. However, in vivo therapeutic effect of PSA scaffolds towards SCI is still lack of evidence and needs to be further explored. In this study, a novel electrospun polycaprolactone/PSA scaffold loaded with methylprednisolone (MP) was developed to achieve efficient therapeutic effects towards SCI. And we believe that it broadens the application of PSA for SCI treatment.
Collapse
|
25
|
Li R, Sahu S, Schachner M. Phenelzine, a small organic compound mimicking the functions of cell adhesion molecule L1, promotes functional recovery after mouse spinal cord injury. Restor Neurol Neurosci 2018; 36:469-483. [PMID: 29889084 DOI: 10.3233/rnn-170808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Neural cell adhesion molecule L1 contributes to nervous system development and maintenance by promoting neuronal survival, neuritogenesis, axonal regrowth/sprouting, myelination, and synapse formation and plasticity. L1 also enhances recovery after spinal cord injury and ameliorates neurodegenerative processes in experimental rodent models. Aiming for clinical translation of L1 into therapy we screened for and functionally characterized in vitro the small organic molecule phenelzine, which mimics characteristic L1 functions. OBJECTIVE The present study was designed to evaluate the potential of this compound in vivo in a mouse model of spinal cord injury. METHODS AND RESULTS In mice, intraperitoneal injection of phenelzine immediately after severe thoracic compression, and thereafter once daily for 6 weeks, improved hind limb function, reduced astrogliosis and promoted axonal regrowth/sprouting at 4 and 5 weeks after spinal cord injury compared to vehicle control-treated mice. Phenelzine application upregulated L1 expression in the spinal cord and stimulated the cognate L1-mediated intracellular signaling cascades in the spinal cord tissue. Phenelzine-treated mice showed decreased levels of pro-inflammatory cytokines, such as interleukin-1β, interleukin-6, and tumor necrosis factor-α in the injured spinal cord during the acute phase of inflammation. CONCLUSIONS This study provides new insights into the role of phenelzine in L1-mediated neural functions and modulation of inflammation. The combined results raise hopes that phenelzine may develop into a therapeutic agent for nervous system injuries.
Collapse
Affiliation(s)
- Rong Li
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| | - Sudhanshu Sahu
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
26
|
Loers G, Astafiev S, Hapiak Y, Saini V, Mishra B, Gul S, Kaur G, Schachner M, Theis T. The polysialic acid mimetics idarubicin and irinotecan stimulate neuronal survival and neurite outgrowth and signal via protein kinase C. J Neurochem 2017; 142:392-406. [PMID: 28542923 PMCID: PMC5539918 DOI: 10.1111/jnc.14076] [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] [Received: 01/30/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 02/05/2023]
Abstract
Polysialic acid (PSA) is a large, negatively charged, linear homopolymer of alpha2-8-linked sialic acid residues. It is generated by two polysialyltransferases and attached to N- and/or O-linked glycans, and its main carrier is the neural cell adhesion molecule (NCAM). PSA controls the development and regeneration of the nervous system by enhancing cell migration, axon pathfinding, synaptic targeting, synaptic plasticity, by regulating the differentiation of progenitor cells and by modulating cell-cell and cell-matrix adhesions. In the adult, PSA plays a role in the immune system, and PSA mimetics promote functional recovery after nervous system injury. In search for novel small molecule mimetics of PSA that are applicable for therapy, we identified idarubicin, an antineoplastic anthracycline, and irinotecan, an antineoplastic agent of the topoisomerase I inhibitor class, as PSA mimetics using a competition enzyme-linked immunosorbent assay. Idarubicin and irinotecan compete with the PSA-mimicking peptide and colominic acid, the bacterial analog of PSA, for binding to the PSA-specific monoclonal antibody 735. Idarubicin and irinotecan stimulate neurite outgrowth and survival of cultured cerebellar neurons after oxidative stress via protein kinase C and Erk1/2 in a similar manner as colominic acid, whereas Fyn, casein kinase II and the phosphatase and tensin homolog are only involved in idarubicin and irinotecan-stimulated neurite outgrowth. These novel results show that the structure and function of PSA can be mimicked by the small organic compounds irinotecan and idarubicin which trigger the same signaling cascades as PSA, thus introducing the possibility of retargeting these drugs to treat nervous system injuries.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Steven Astafiev
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Yuliya Hapiak
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Vedangana Saini
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port (Fraunhofer-IME SP), Schnackenburgalle114, D-22525 Hamburg, Germany
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
- To whom correspondence should be addressed: Melitta Schachner, Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA; phone: +1-732-445-1780; fax: +1-732-445-2063; ; or Melitta Schachner, Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China; phone: + 86 754 8890 0276; fax: + 86 754 8890 0236;
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| |
Collapse
|
27
|
Ieronymaki M, Nuti F, Brancaccio D, Rossi G, Real-Fernández F, Cao Y, Monasson O, Larregola M, Peroni E, Uziel J, Sabatino G, Novellino E, Carotenuto A, Papini AM, Rovero P. Structure-Activity Relationship Studies, SPR Affinity Characterization, and Conformational Analysis of Peptides That Mimic the HNK-1 Carbohydrate Epitope. ChemMedChem 2017; 12:751-759. [PMID: 28403522 DOI: 10.1002/cmdc.201700042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/23/2017] [Indexed: 01/08/2023]
Abstract
The design of molecules that mimic biologically relevant glycans is a significant goal for understanding important biological processes and may lead to new therapeutic and diagnostic agents. In this study we focused our attention on the trisaccharide human natural killer cell-1 (HNK-1), considered the antigenic determinant of myelin-associated glycoprotein and the target of clinically relevant auto-antibodies in autoimmune neurological disorders such as IgM monoclonal gammopathy and demyelinating polyneuropathy. We describe a structure-activity relationship study based on surface plasmon resonance binding affinities aimed at the optimization of a peptide that mimics the HNK-1 minimal epitope. We developed a cyclic heptapeptide that shows an affinity of 1.09×10-7 m for a commercial anti-HNK1 mouse monoclonal antibody. Detailed conformational analysis gave possible explanations for the good affinity displayed by this novel analogue, which was subsequently used as an immunological probe. However, preliminary screening indicates that patients' sera do not specifically recognize this peptide, showing that murine monoclonal antibodies cannot be used as a guide to select immunological probes for the detection of clinically relevant human auto-antibodies.
Collapse
Affiliation(s)
- Matthaia Ieronymaki
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy
| | - Francesca Nuti
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy
| | - Diego Brancaccio
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Napoli, Italy
| | - Giada Rossi
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical Sciences and Nutraceutics, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Feliciana Real-Fernández
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical Sciences and Nutraceutics, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Yihong Cao
- PeptLab, UCP Platform and Laboratory of Chemical Biology EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac, 95031, Cergy-Pontoise Cedex, France
| | - Olivier Monasson
- PeptLab, UCP Platform and Laboratory of Chemical Biology EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac, 95031, Cergy-Pontoise Cedex, France
| | - Maud Larregola
- PeptLab, UCP Platform and Laboratory of Chemical Biology EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac, 95031, Cergy-Pontoise Cedex, France
| | - Elisa Peroni
- PeptLab, UCP Platform and Laboratory of Chemical Biology EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac, 95031, Cergy-Pontoise Cedex, France
| | - Jacques Uziel
- PeptLab, UCP Platform and Laboratory of Chemical Biology EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac, 95031, Cergy-Pontoise Cedex, France
| | - Giuseppina Sabatino
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Napoli, Italy
| | - Alfonso Carotenuto
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Napoli, Italy
| | - Anna Maria Papini
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,PeptLab, UCP Platform and Laboratory of Chemical Biology EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac, 95031, Cergy-Pontoise Cedex, France
| | - Paolo Rovero
- Laboratory of Peptide and Protein Chemistry and Biology, PeptLab, Via della Lastruccia 13, 50019, Sesto Fiorentino, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical Sciences and Nutraceutics, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
28
|
Wuttke S, Zimpel A, Bein T, Braig S, Stoiber K, Vollmar A, Müller D, Haastert-Talini K, Schaeske J, Stiesch M, Zahn G, Mohmeyer A, Behrens P, Eickelberg O, Bölükbas DA, Meiners S. Validating Metal-Organic Framework Nanoparticles for Their Nanosafety in Diverse Biomedical Applications. Adv Healthc Mater 2017; 6. [PMID: 27863166 DOI: 10.1002/adhm.201600818] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/06/2016] [Indexed: 01/09/2023]
Abstract
Metal-organic frameworks (MOFs) are promising platforms for the synthesis of nanoparticles for diverse medical applications. Their fundamental design principles allow for significant control of the framework architecture and pore chemistry, enabling directed functionalization for nanomedical applications. However, before applying novel nanomaterials to patients, it is imperative to understand their potential health risks. In this study, the nanosafety of different MOF nanoparticles is analyzed comprehensively for diverse medical applications. The authors first evaluate the effects of MOFs on human endothelial and mouse lung cells, which constitute a first line of defense upon systemic blood-mediated and local lung-specific applications of nanoparticles. Second, we validated these MOFs for multifunctional surface coatings of dental implants using human gingiva fibroblasts. Moreover, biocompatibility of MOFs is assessed for surface coating of nerve guidance tubes using human Schwann cells and rat dorsal root ganglion cultures. The main finding of this study is that the nanosafety and principal suitability of our MOF nanoparticles as novel agents for drug delivery and implant coatings strongly varies with the effector cell type. We conclude that it is therefore necessary to carefully evaluate the nanosafety of MOF nanomaterials with respect to their particular medical application and their interacting primary cell types, respectively.
Collapse
Affiliation(s)
- Stefan Wuttke
- Department of Chemistry and Center for NanoScience (CeNS); University of Munich (LMU); 81377 Munich Germany
| | - Andreas Zimpel
- Department of Chemistry and Center for NanoScience (CeNS); University of Munich (LMU); 81377 Munich Germany
| | - Thomas Bein
- Department of Chemistry and Center for NanoScience (CeNS); University of Munich (LMU); 81377 Munich Germany
| | - Simone Braig
- Department of Pharmacy; University of Munich (LMU); 81377 Munich Germany
| | - Katharina Stoiber
- Department of Pharmacy; University of Munich (LMU); 81377 Munich Germany
| | - Angelika Vollmar
- Department of Pharmacy; University of Munich (LMU); 81377 Munich Germany
| | - Dominik Müller
- Institute of Neuroanatomy; Hannover Medical School; Hannover 30625 Germany
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy; Hannover Medical School; Hannover 30625 Germany
- Center for Systems Neurosciences (ZSN) Hannover; 30625 Hannover Germany
| | - Jörn Schaeske
- Department of Prosthetic Dentistry and Biomedical Materials Science; Hannover Medical School; Hannover 30625 Germany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science; Hannover Medical School; Hannover 30625 Germany
| | - Gesa Zahn
- Institute for Inorganic Chemistry; Leibniz University Hannover; Hannover 30167 Germany
| | - Alexander Mohmeyer
- Institute for Inorganic Chemistry; Leibniz University Hannover; Hannover 30167 Germany
| | - Peter Behrens
- Institute for Inorganic Chemistry; Leibniz University Hannover; Hannover 30167 Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center (CPC); University Hospital, University of Munich (LMU); Member of the German Center for Lung Research (DZL); 81377 Munich Germany
| | - Deniz A. Bölükbas
- Comprehensive Pneumology Center (CPC); University Hospital, University of Munich (LMU); Member of the German Center for Lung Research (DZL); 81377 Munich Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC); University Hospital, University of Munich (LMU); Member of the German Center for Lung Research (DZL); 81377 Munich Germany
| |
Collapse
|
29
|
Wang XJ, Gao YP, Lu NN, Li WS, Xu JF, Ying XY, Wu G, Liao MH, Tan C, Shao LX, Lu YM, Zhang C, Fukunaga K, Han F, Du YZ. Endogenous Polysialic Acid Based Micelles for Calmodulin Antagonist Delivery against Vascular Dementia. ACS APPLIED MATERIALS & INTERFACES 2016; 8:35045-35058. [PMID: 27750011 DOI: 10.1021/acsami.6b13052] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Clinical treatment for vascular dementia still remains a challenge mainly due to the blood-brain barrier (BBB). Here, a micelle based on polysialic acid (PSA), which is a hydrophilic and endogenous carbohydrate polymer, was designed to deliver calmodulin antagonist for therapy of vascular dementia. PSA was first chemically conjugated with octadecylamine (ODA), and the obtained PSA-ODA copolymer could self-assemble into micelle in aqueous solution with a 120.0 μg/mL critical micelle concentration. The calmodulin antagonist loaded PSA-ODA micelle, featuring sustained drug release behavior over a period of 72 h with a 3.6% (w/w) drug content and a 107.0 ± 4.0 nm size was then fabricated. The PSA-ODA micelle could cross the BBB mainly via active endocytosis by brain endothelial cells followed by transcytosis. In a water maze test for spatial learning, calmodulin antagonist loaded PSA-ODA micelle significantly reduced the escape latencies of right unilateral common carotid arteries occlusion (rUCCAO) mice with dosage significantly reduced versus free drug. The decrease of hippocampal phospho-CaMKII (Thr286/287) and phospho-synapsin I (Ser603) was partially restored in rUCCAO mice following calmodulin antagonist loaded PSA-ODA micelle treatment. Consistent with the restored CaMKII phosphorylation, the elevation of BrdU/NeuN double-positive cells in the same context was also observed. Overall, the PSA-ODA micelle developed from the endogenous material might promote the development of therapeutic approaches for improving the efficacy of brain-targeted drug delivery and have great potential for vascular dementia treatment.
Collapse
Affiliation(s)
| | - Yin-Ping Gao
- School of Medicine, Zhejiang University City College , Hangzhou 310058, China
| | | | | | | | | | | | | | | | | | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College , Hangzhou 310058, China
| | | | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University , Sendai 980-8574, Japan
| | | | | |
Collapse
|
30
|
Kataria H, Lutz D, Chaudhary H, Schachner M, Loers G. Small Molecule Agonists of Cell Adhesion Molecule L1 Mimic L1 Functions In Vivo. Mol Neurobiol 2016; 53:4461-83. [PMID: 26253722 DOI: 10.1007/s12035-015-9352-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/07/2015] [Indexed: 02/05/2023]
Abstract
Lack of permissive mechanisms and abundance of inhibitory molecules in the lesioned central nervous system of adult mammals contribute to the failure of functional recovery after injury, leading to severe disabilities in motor functions and pain. Peripheral nerve injury impairs motor, sensory, and autonomic functions, particularly in cases where nerve gaps are large and chronic nerve injury ensues. Previous studies have indicated that the neural cell adhesion molecule L1 constitutes a viable target to promote regeneration after acute injury. We screened libraries of known drugs for small molecule agonists of L1 and evaluated the effect of hit compounds in cell-based assays in vitro and in mice after femoral nerve and spinal cord injuries in vivo. We identified eight small molecule L1 agonists and showed in cell-based assays that they stimulate neuronal survival, neuronal migration, and neurite outgrowth and enhance Schwann cell proliferation and migration and myelination of neurons in an L1-dependent manner. In a femoral nerve injury mouse model, enhanced functional regeneration and remyelination after application of the L1 agonists were observed. In a spinal cord injury mouse model, L1 agonists improved recovery of motor functions, being paralleled by enhanced remyelination, neuronal survival, and monoaminergic innervation, reduced astrogliosis, and activation of microglia. Together, these findings suggest that application of small organic compounds that bind to L1 and stimulate the beneficial homophilic L1 functions may prove to be a valuable addition to treatments of nervous system injuries.
Collapse
Affiliation(s)
- Hardeep Kataria
- Institut für Biosynthese Neuraler Strukturen, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum-Hamburg Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - David Lutz
- Institut für Biosynthese Neuraler Strukturen, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum-Hamburg Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Harshita Chaudhary
- Institut für Biosynthese Neuraler Strukturen, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum-Hamburg Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
| | - Gabriele Loers
- Institut für Biosynthese Neuraler Strukturen, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum-Hamburg Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| |
Collapse
|
31
|
Lutz D, Kataria H, Kleene R, Loers G, Chaudhary H, Guseva D, Wu B, Jakovcevski I, Schachner M. Myelin Basic Protein Cleaves Cell Adhesion Molecule L1 and Improves Regeneration After Injury. Mol Neurobiol 2016; 53:3360-3376. [PMID: 26081148 DOI: 10.1007/s12035-015-9277-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/01/2015] [Indexed: 02/05/2023]
Abstract
Myelin basic protein (MBP) is a serine protease that cleaves neural cell adhesion molecule L1 and generates a transmembrane L1 fragment which facilitates L1-dependent functions in vitro, such as neurite outgrowth, neuronal cell migration and survival, myelination by Schwann cells as well as Schwann cell proliferation, migration, and process formation. Ablation and blocking of MBP or disruption of its proteolytic activity by mutation of a proteolytically active serine residue abolish L1-dependent cellular responses. In utero injection of adeno-associated virus encoding proteolytically active MBP into MBP-deficient shiverer mice normalizes differentiation, myelination, and synaptogenesis in the developing postnatal spinal cord, in contrast to proteolytically inactive MBP. Application of active MBP to the injured wild-type spinal cord and femoral nerve augments levels of a transmembrane L1 fragment, promotes remyelination, and improves functional recovery after injury. Application of MBP antibody impairs recovery. Virus-mediated expression of active MBP in the lesion site after spinal cord injury results in improved functional recovery, whereas injection of virus encoding proteolytically inactive MBP fails to do so. The present study provides evidence for a novel L1-mediated function of MBP in the developing spinal cord and in the injured adult mammalian nervous system that leads to enhanced recovery after acute trauma.
Collapse
Affiliation(s)
- David Lutz
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Hardeep Kataria
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Harshita Chaudhary
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Daria Guseva
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Cellular Neurobiology, Medical School Hannover, Hannover, Germany
| | - Bin Wu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Igor Jakovcevski
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melitta Schachner
- Melitta Schachner, Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
32
|
Saini V, Lutz D, Kataria H, Kaur G, Schachner M, Loers G. The polysialic acid mimetics 5-nonyloxytryptamine and vinorelbine facilitate nervous system repair. Sci Rep 2016; 6:26927. [PMID: 27324620 PMCID: PMC4914991 DOI: 10.1038/srep26927] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/07/2016] [Indexed: 02/05/2023] Open
Abstract
Polysialic acid (PSA) is a large negatively charged glycan mainly attached to the neural cell adhesion molecule (NCAM). Several studies have shown that it is important for correct formation of brain circuitries during development and for synaptic plasticity, learning and memory in the adult. PSA also plays a major role in nervous system regeneration following injury. As a next step for clinical translation of PSA based therapeutics, we have previously identified the small organic compounds 5-nonyloxytryptamine and vinorelbine as PSA mimetics. Activity of 5-nonyloxytryptamine and vinorelbine had been confirmed in assays with neural cells from the central and peripheral nervous system in vitro and shown to be independent of their function as serotonin receptor 5-HT1B/1D agonist or cytostatic drug, respectively. As we show here in an in vivo paradigm for spinal cord injury in mice, 5-nonyloxytryptamine and vinorelbine enhance regain of motor functions, axonal regrowth, motor neuron survival and remyelination. These data indicate that 5-nonyloxytryptamine and vinorelbine may be re-tasked from their current usage as a 5-HT1B/1D agonist or cytostatic drug to act as mimetics for PSA to stimulate regeneration after injury in the mammalian nervous system.
Collapse
Affiliation(s)
- Vedangana Saini
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - David Lutz
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Hardeep Kataria
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Keck Center for Collaborative Neurosciences, Rutgers University, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, People’s Republic of China
| | - Gabriele Loers
- Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| |
Collapse
|
33
|
Zhang R, Loers G, Schachner M, Boelens R, Wienk H, Siebert S, Eckert T, Kraan S, Rojas-Macias MA, Lütteke T, Galuska SP, Scheidig A, Petridis AK, Liang S, Billeter M, Schauer R, Steinmeyer J, Schröder JM, Siebert HC. Molecular Basis of the Receptor Interactions of Polysialic Acid (polySia), polySia Mimetics, and Sulfated Polysaccharides. ChemMedChem 2016; 11:990-1002. [PMID: 27136597 DOI: 10.1002/cmdc.201500609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/01/2016] [Indexed: 02/05/2023]
Abstract
Polysialic acid (polySia) and polySia glycomimetic molecules support nerve cell regeneration, differentiation, and neuronal plasticity. With a combination of biophysical and biochemical methods, as well as data mining and molecular modeling techniques, it is possible to correlate specific ligand-receptor interactions with biochemical processes and in vivo studies that focus on the potential therapeutic impact of polySia, polySia glycomimetics, and sulfated polysaccharides in neuronal diseases. With this strategy, the receptor interactions of polySia and polySia mimetics can be understood on a submolecular level. As the HNK-1 glycan also enhances neuronal functions, we tested whether similar sulfated oligo- and polysaccharides from seaweed could be suitable, in addition to polySia, for finding potential new routes into patient care focusing on an improved cure for various neuronal diseases. The knowledge obtained here on the structural interplay between polySia or sulfated polysaccharides and their receptors can be exploited to develop new drugs and application routes for the treatment of neurological diseases and dysfunctions.
Collapse
Affiliation(s)
- Ruiyan Zhang
- RI-B-NT: Research Institute of Bioinformatics and Nanotechnology, Franziusallee 177, 24148, Kiel, Germany
- Zoological Institute, Department of Structural Biology, Kiel University, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Falkenried 94, 20251, Hamburg, Germany
| | - Melitta Schachner
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Falkenried 94, 20251, Hamburg, Germany
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Rolf Boelens
- Bijvoet Center for Biomolecular Research, NMR Spectroscopy, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hans Wienk
- Bijvoet Center for Biomolecular Research, NMR Spectroscopy, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Simone Siebert
- RI-B-NT: Research Institute of Bioinformatics and Nanotechnology, Franziusallee 177, 24148, Kiel, Germany
| | - Thomas Eckert
- Institute of Veterinary Physiology and Biochemistry, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Gießen, Frankfurter Str. 100, 35392, Gießen, Germany
- Clinic for Obstetrics, Gynecology and Andrology of Large and Small Animals, Justus-Liebig-Universität Gießen, Frankfurter Str. 106, 35392, Gießen, Germany
| | - Stefan Kraan
- Ocean Harvest Technology Ltd., N17 Business Park, Milltown, County Galway, Ireland
| | - Miguel A Rojas-Macias
- Institute of Veterinary Physiology and Biochemistry, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Gießen, Frankfurter Str. 100, 35392, Gießen, Germany
| | - Thomas Lütteke
- Institute of Veterinary Physiology and Biochemistry, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Gießen, Frankfurter Str. 100, 35392, Gießen, Germany
| | - Sebastian P Galuska
- Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-Universität Gießen, Friedrichstr. 24, 35392, Gießen, Germany
| | - Axel Scheidig
- Zoological Institute, Department of Structural Biology, Kiel University, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Athanasios K Petridis
- Neurosurgery Clinic, University Düsseldorf, Moorenstraße 5, 40255, Düsseldorf, Germany
| | - Songping Liang
- College of Life Sciences, Hunan Normal University, 410081, Changsha, China
| | - Martin Billeter
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 100, 40530, Gothenburg, Sweden
| | - Roland Schauer
- Institute of Biochemistry, Kiel University, Olshausenstr. 40, 24098, Kiel, Germany
| | - Jürgen Steinmeyer
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University Hospital Giessen and Marburg GmbH, Paul-Meimberg-Str. 3, 35392, Gießen, Germany
| | - Jens-Michael Schröder
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, 24105, Kiel, Germany
| | - Hans-Christian Siebert
- RI-B-NT: Research Institute of Bioinformatics and Nanotechnology, Franziusallee 177, 24148, Kiel, Germany.
| |
Collapse
|
34
|
Ezra M, Bushman J, Shreiber D, Schachner M, Kohn J. Porous and Nonporous Nerve Conduits: The Effects of a Hydrogel Luminal Filler With and Without a Neurite-Promoting Moiety. Tissue Eng Part A 2016; 22:818-26. [PMID: 27102571 PMCID: PMC4876540 DOI: 10.1089/ten.tea.2015.0354] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 04/21/2016] [Indexed: 02/05/2023] Open
Abstract
Nerve conduits prefilled with hydrogels are frequently explored in an attempt to promote nerve regeneration. This study examines the interplay in vivo between the porosity of the conduit wall and the level of bioactivity of the hydrogel used to fill the conduit. Nerve regeneration in porous (P) or nonporous (NP) conduits that were filled with either collagen only or collagen enhanced with a covalently attached neurite-promoting peptide mimic of the glycan human natural killer cell antigen-1 (m-HNK) were compared in a 5 mm critical size defect in the mouse femoral nerve repair model. Although collagen is a cell-friendly matrix that does not differentiate between neural and nonneural cells, the m-HNK-enhanced collagen specifically promotes axon growth and appropriate motor neuron targeting. In this study, animals treated with NP conduits filled with collagen grafted with m-HNK (CollagenHNK) had the best overall functional recovery, based on a range of histomorphometric observations and parameters of functional recovery. Our data indicate that under some conditions, the use of generally cell friendly fillers such as collagen may limit nerve regeneration. This finding is significant, considering the frequent use of collagen-based hydrogels as fillers of nerve conduits.
Collapse
Affiliation(s)
- Mindy Ezra
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Jared Bushman
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - David Shreiber
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Melitta Schachner
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
35
|
Lee YS, Griffin J, Masand SN, Shreiber DI, Uhrich KE. Salicylic acid-based poly(anhydride-ester) nerve guidance conduits: Impact of localized drug release on nerve regeneration. J Biomed Mater Res A 2016; 104:975-82. [PMID: 26691691 DOI: 10.1002/jbm.a.35630] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/05/2015] [Accepted: 12/16/2015] [Indexed: 11/10/2022]
Abstract
Nerve guidance conduits (NGCs) can serve as physical scaffolds aligning and supporting regenerating cells while preventing scar tissue formation that often interferes with the regeneration process. Numerous studies have focused on functionalizing NGCs with neurotrophic factors, for example, to support nerve regeneration over longer gaps, but few directly incorporate therapeutic agents. Herein, we fabricated NGCs from a polyanhydride comprised of salicylic acid (SA), a nonsteroidal anti-inflammatory drug, then performed in vitro and in vivo assays. In vitro studies included cytotoxicity, anti-inflammatory response, and NGC porosity measurements. To prepare for implantation, type I collagen hydrogels were used as NGC luminal fillers to further enhance the axonal regeneration process. For the in vivo studies, SA-NGCs were implanted in femoral nerves of mice for 16 weeks and evaluated for functional recovery. The SA-based NGCs functioned as both a drug delivery vehicle capable of reducing inflammation and scar tissue formation because of SA release as well as a tissue scaffold that promotes peripheral nerve regeneration and functional recovery.
Collapse
Affiliation(s)
- Yong S Lee
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854
| | - Jeremy Griffin
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854
| | - Shirley N Masand
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854
| | - Kathryn E Uhrich
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854.,Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854
| |
Collapse
|
36
|
Loers G, Saini V, Mishra B, Gul S, Chaudhury S, Wallqvist A, Kaur G, Schachner M. Vinorelbine and epirubicin share common features with polysialic acid and modulate neuronal and glial functions. J Neurochem 2016; 136:48-62. [PMID: 26443186 PMCID: PMC4904230 DOI: 10.1111/jnc.13383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 02/05/2023]
Abstract
Polysialic acid (PSA), a large, linear glycan composed of 8 to over 100 α2,8-linked sialic acid residues, modulates development of the nervous system by enhancing cell migration, axon pathfinding, and synaptic targeting and by regulating differentiation of progenitor cells. PSA also functions in developing and adult immune systems and is a signature of many cancers. In this study we identified vinorelbine, a semi-synthetic third generation vinca alkaloid, and epirubicin, an anthracycline and 4'-epimer of doxorubicin, as PSA mimetics. Similar to PSA, vinorelbine and epirubicin bind to the PSA-specific monoclonal antibody 735 and compete with the bacterial analog of PSA, colominic acid in binding to monoclonal antibody 735. Vinorelbine and epirubicin stimulate neurite outgrowth of cerebellar neurons via the neural cell adhesion molecule, via myristoylated alanine-rich C kinase substrate, and via fibroblast growth factor receptor, signaling through Erk pathways. Furthermore, the two compounds enhance process formation of Schwann cells and migration of cerebellar neurons in culture, and reduce migration of astrocytes after injury. These novel results show that the structure and function of PSA can be mimicked by the small organic compounds vinorelbine and epirubicin, thus raising the possibility to re-target drugs used in treatment of cancers to nervous system repair. Vinorelbine and epirubicin, identified as PSA mimetics, enhance, like PSA, neuronal migration, neuritogenesis, and formation of Schwann cell processes, and reduce astrocytic migration. Ablating NCAM, inhibiting fibroblast growth factor (FGFR) receptor, or adding the effector domain of myristoylated alanine-rich C kinase substrate (MARCKS) minimize the vinorelbine and epirubicin effects, indicating that they are true PSA mimetics triggering PSA-mediated functions.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Vedangana Saini
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology ScreeningPort (Fraunhofer-IME SP), Schnackenburgalle114, D-22525 Hamburg, Germany
| | - Sidhartha Chaudhury
- DoD Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Materiel Command, Fort Detrick, MD 21702 (USA)
| | - Anders Wallqvist
- DoD Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Materiel Command, Fort Detrick, MD 21702 (USA)
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| |
Collapse
|
37
|
Functionalized composite scaffolds improve the engraftment of transplanted dopaminergic progenitors in a mouse model of Parkinson's disease. Biomaterials 2016; 74:89-98. [DOI: 10.1016/j.biomaterials.2015.09.039] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 12/16/2022]
|
38
|
Chen H, Liu C, Yin J, Chen Z, Xu J, Wang D, Zhu J, Zhang Z, Sun Y, Li A. Mitochondrial Cyclophilin D as a Potential Therapeutic Target for Ischemia-Induced Facial Palsy in Rats. Cell Mol Neurobiol 2015; 35:931-41. [PMID: 25820785 DOI: 10.1007/s10571-015-0188-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
Abstract
Many studies have demonstrated that ischemia could induce facial nerve (FN) injury. However, there is a lack of a suitable animal model for FN injury study and thus little knowledge is available about the precise mechanism for FN injury. The aims of this study were to establish a reliable FN injury model induced by blocking the petrosal artery and to investigate whether dysfunctional interaction between cyclophilin D (CypD) and mitochondrial permeability transition pore (MPTP) can mediate cell dysfunction in ischemic FN injury. The outcomes of ischemia-induced FN injury rat model were evaluated by behavioral assessment, histological observation, electrophysiology, and electron microscopy. Then the levels of CypD and protein that forms the MPTP were evaluated under the conditions with or without the treatment of Cyclosporin A (CsA), which has been found to disrupt MPTP through the binding of CypD. The blocking of petrosal artery caused significant facial palsy signs in the ischemia group but not in the sham group. Furthermore, ischemia can induce the dysfunction of facial nucleus neurons and destruction of the myelin sheath and increase the protein levels of CypD and MPTP protein compared with sham group. Interestingly, treatment with CsA significantly improved neurological function and reversed the ischemia-induced increase of CypD and MPTP proteins in ischemia group. These results demonstrated that blocking of petrosal artery in rats can induce FN injury and the mechanism may be related to the disruption of MPTP by CypD.
Collapse
Affiliation(s)
- Huizhen Chen
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Chnagtao Liu
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Jie Yin
- Department of Neurosurgery, Xuzhou Central Hospital, Xuzhou, Jiangsu, 221009, People's Republic of China
| | - Zhen Chen
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Jinwang Xu
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Duanlei Wang
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Jiaqiu Zhu
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Ziyuan Zhang
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Yong Sun
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China
| | - Aimin Li
- Department of Neurosurgery, The First People's Hospital of Lianyungang, 182 Tong Guan North Road, Lianyungang, Jiangsu, 222002, People's Republic of China.
| |
Collapse
|
39
|
Rohrbeck A, Stahl F, Höltje M, Hettwer T, Lindner P, Hagemann S, Pich A, Haastert-Talini K. C3-induced release of neurotrophic factors from Schwann cells - potential mechanism behind its regeneration promoting activity. Neurochem Int 2015; 90:232-45. [PMID: 26417907 DOI: 10.1016/j.neuint.2015.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/11/2015] [Accepted: 09/23/2015] [Indexed: 01/10/2023]
Abstract
Previous studies revealed a peripheral nerve regeneration (PNR)(1) promoting activity of Clostridium botulinum C3(2) exoenzyme or a 26(mer) C-terminal peptide fragment covering amino acids 156-181 (C3(156-181)),(3) when delivered as one-time injection at the lesion site. The current study was performed to 1) investigate if prolonged availability of C3 and C3(156-181) at the lesion site can further enhance PNR in vivo and to 2) elucidate effects of C3 and C3(156-181) on Schwann cells (SCs)(4)in vitro. For in vivo studies, 10 mm adult rat sciatic nerve gaps were reconstructed with the epineurial pouch technique or autologous nerve grafts. Epineurial pouches were filled with a hydrogel containing i) vehicle, ii) 40 μM C3 or iii) 40 μM C3(156-181). Sensory and motor functional recovery was monitored over 12 weeks and the outcome of PNR further analyzed by nerve morphometry. In vitro, we compared gene expression profiles (microarray analysis) and neurotrophic factor expression (western blot analysis) of untreated rat neonatal SCs with those treated with C3 or C3(156-181) for 72 h. Effects on neurotrophic factor expression levels were proven in adult human SCs. Unexpectedly, prolonged delivery of C3 and C3(156-181) at the lesion site did not increase the outcome of PNR. Regarding the potential mechanism underlying their previously detected PNR promoting action, however, 6 genes were found to be commonly altered in SCs upon treatment with C3 or C3(156-181). We demonstrate significant down-regulation of genes involved in glutamate uptake (Eaac1,(5)Grin2a(6)) and changes in neurotrophic factor expression (increase of FGF-2(7) and decrease of NGF(8)). Our microarray-based expression profiling revealed novel C3-regulated genes in SCs possibly involved in the axonotrophic (regeneration promoting) effects of C3 and C3(156-181). Detection of altered neurotrophic factor expression by C3 or C3(156-181) treated primary neonatal rat SCs and primary adult human SCs supports this hypothesis.
Collapse
Affiliation(s)
- Astrid Rohrbeck
- Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.
| | - Frank Stahl
- Institute for Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, D-30167 Hannover, Germany
| | - Markus Höltje
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | - Timo Hettwer
- Institute of Neuroanatomy, Hannover Medical School and Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Patrick Lindner
- Institute for Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, D-30167 Hannover, Germany
| | - Sandra Hagemann
- Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy, Hannover Medical School and Carl-Neuberg-Str. 1, D-30625 Hannover, Germany; Center for Systems Neuroscience (ZSN), Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.
| |
Collapse
|
40
|
Mehrabian M, Brethour D, Wang H, Xi Z, Rogaeva E, Schmitt-Ulms G. The Prion Protein Controls Polysialylation of Neural Cell Adhesion Molecule 1 during Cellular Morphogenesis. PLoS One 2015; 10:e0133741. [PMID: 26288071 PMCID: PMC4546001 DOI: 10.1371/journal.pone.0133741] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/19/2015] [Indexed: 01/06/2023] Open
Abstract
Despite its multi-faceted role in neurodegenerative diseases, the physiological function of the prion protein (PrP) has remained elusive. On the basis of its evolutionary relationship to ZIP metal ion transporters, we considered that PrP may contribute to the morphogenetic reprogramming of cells underlying epithelial-to-mesenchymal transitions (EMT). Consistent with this hypothesis, PrP transcription increased more than tenfold during EMT, and stable PrP-deficient cells failed to complete EMT in a mammalian cell model. A global comparative proteomics analysis identified the neural cell adhesion molecule 1 (NCAM1) as a candidate mediator of this impairment, which led to the observation that PrP-deficient cells fail to undergo NCAM1 polysialylation during EMT. Surprisingly, this defect was caused by a perturbed transcription of the polysialyltransferase ST8SIA2 gene. Proteomics data pointed toward β-catenin as a transcriptional regulator affected in PrP-deficient cells. Indeed, pharmacological blockade or siRNA-based knockdown of β-catenin mimicked PrP-deficiency in regards to NCAM1 polysialylation. Our data established the existence of a PrP-ST8SIA2-NCAM signaling loop, merged two mature fields of investigation and offer a simple model for explaining phenotypes linked to PrP.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dylan Brethour
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hansen Wang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Zhengrui Xi
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
41
|
Rowlands D, Sugahara K, Kwok JCF. Glycosaminoglycans and glycomimetics in the central nervous system. Molecules 2015; 20:3527-48. [PMID: 25706756 PMCID: PMC6272379 DOI: 10.3390/molecules20033527] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/09/2015] [Accepted: 02/13/2015] [Indexed: 01/05/2023] Open
Abstract
With recent advances in the construction of synthetic glycans, selective targeting of the extracellular matrix (ECM) as a potential treatment for a wide range of diseases has become increasingly popular. The use of compounds that mimic the structure or bioactive function of carbohydrate structures has been termed glycomimetics. These compounds are mostly synthetic glycans or glycan-binding constructs which manipulate cellular interactions. Glycosaminoglycans (GAGs) are major components of the ECM and exist as a diverse array of differentially sulphated disaccharide units. In the central nervous system (CNS), they are expressed by both neurons and glia and are crucial for brain development and brain homeostasis. The inherent diversity of GAGs make them an essential biological tool for regulating a complex range of cellular processes such as plasticity, cell interactions and inflammation. They are also involved in the pathologies of various neurological disorders, such as glial scar formation and psychiatric illnesses. It is this diversity of functions and potential for selective interventions which makes GAGs a tempting target. In this review, we shall describe the molecular make-up of GAGs and their incorporation into the ECM of the CNS. We shall highlight the different glycomimetic strategies that are currently being used in the nervous system. Finally, we shall discuss some possible targets in neurological disorders that may be addressed using glycomimetics.
Collapse
Affiliation(s)
- Dáire Rowlands
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK.
| | - Kazuyuki Sugahara
- Proteoglycan Signaling and Therapeutics Research Group, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan.
| | - Jessica C F Kwok
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK.
| |
Collapse
|
42
|
Struzyna LA, Katiyar K, Cullen DK. Living scaffolds for neuroregeneration. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2014; 18:308-318. [PMID: 28736499 PMCID: PMC5520662 DOI: 10.1016/j.cossms.2014.07.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Neural tissue engineers are exploiting key mechanisms responsible for neural cell migration and axonal path finding during embryonic development to create living scaffolds for neuroregeneration following injury and disease. These mechanisms involve the combined use of haptotactic, chemotactic, and mechanical cues to direct cell movement and re-growth. Living scaffolds provide these cues through the use of cells engineered in a predefined architecture, generally in combination with biomaterial strategies. Although several hurdles exist in the implementation of living regenerative scaffolds, there are considerable therapeutic advantages to using living cells in conjunction with biomaterials. The leading contemporary living scaffolds for neurorepair are utilizing aligned glial cells and neuronal/axonal tracts to direct regenerating axons across damaged tissue to appropriate targets, and in some cases to directly replace the function of lost cells. Future advances in technology, including the use of exogenous stimulation and genetically engineered stem cells, will further the potential of living scaffolds and drive a new era of personalized medicine for neuroregeneration.
Collapse
Affiliation(s)
- Laura A Struzyna
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Kritika Katiyar
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- School of Biomedical Engineering, Drexel University, Philadelphia, PA, United States
| | - D Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, United States
| |
Collapse
|
43
|
Colley KJ, Kitajima K, Sato C. Polysialic acid: biosynthesis, novel functions and applications. Crit Rev Biochem Mol Biol 2014; 49:498-532. [PMID: 25373518 DOI: 10.3109/10409238.2014.976606] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As an anti-adhesive, a reservoir for key biological molecules, and a modulator of signaling, polysialic acid (polySia) is critical for nervous system development and maintenance, promotes cancer metastasis, tissue regeneration and repair, and is implicated in psychiatric diseases. In this review, we focus on the biosynthesis and functions of mammalian polySia, and the use of polySia in therapeutic applications. PolySia modifies a small subset of mammalian glycoproteins, with the neural cell adhesion molecule, NCAM, serving as its major carrier. Studies show that mammalian polysialyltransferases employ a unique recognition mechanism to limit the addition of polySia to a select group of proteins. PolySia has long been considered an anti-adhesive molecule, and its impact on cell adhesion and signaling attributed directly to this property. However, recent studies have shown that polySia specifically binds neurotrophins, growth factors, and neurotransmitters and that this binding depends on chain length. This work highlights the importance of considering polySia quality and quantity, and not simply its presence or absence, as its various roles are explored. The capsular polySia of neuroinvasive bacteria allows these organisms to evade the host immune response. While this "stealth" characteristic has made meningitis vaccine development difficult, it has also made polySia a worthy replacement for polyetheylene glycol in the generation of therapeutic proteins with low immunogenicity and improved circulating half-lives. Bacterial polysialyltransferases are more promiscuous than the protein-specific mammalian enzymes, and new studies suggest that these enzymes have tremendous therapeutic potential, especially for strategies aimed at neural regeneration and tissue repair.
Collapse
Affiliation(s)
- Karen J Colley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, IL , USA and
| | | | | |
Collapse
|
44
|
Mehanna A, Szpotowicz E, Schachner M, Jakovcevski I. Improved regeneration after femoral nerve injury in mice lacking functional T- and B-lymphocytes. Exp Neurol 2014; 261:147-55. [PMID: 24967682 DOI: 10.1016/j.expneurol.2014.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/18/2014] [Accepted: 06/15/2014] [Indexed: 02/05/2023]
Abstract
The immune system plays important functional roles in regeneration after injury to the mammalian central and peripheral nervous systems. After damage to the peripheral nerve several types of immune cells, invade the nerve within hours after the injury. To gain insights into the contribution of T- and B-lymphocytes to recovery from injury we used the mouse femoral nerve injury paradigm. RAG2-/- mice lacking mature T- and B-lymphocytes due to deletion of the recombination activating gene 2 were subjected to resection and surgical reconstruction of the femoral nerve, with the wild-type mice of the same inbred genetic background serving as controls. According to single frame motion analyses, RAG2-/- mice showed better motor recovery in comparison to control mice at four and eight weeks after injury. Retrograde tracing of regrown/sprouted axons of spinal motoneurons showed increased numbers of correctly projecting motoneurons in the lumbar spinal cord of RAG2-/- mice compared with controls. Whereas there was no difference in the motoneuron soma size between genotypes, RAG2-/- mice displayed fewer cholinergic and inhibitory synaptic terminals around somata of spinal motoneurons both prior to and after injury, compared with wild-type mice. Extent of myelination of regrown axons in the motor branch of the femoral nerve measured as g-ratio was more extensive in RAG2-/- than in control mice eight weeks after injury. We conclude that activated T- and B-lymphocytes restrict motor recovery after femoral nerve injury, associated with the increased survival of motoneurons and improved remyelination.
Collapse
Affiliation(s)
- Ali Mehanna
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; Lebanese International University School of Arts & Sciences, P.O. Box: 146404 Mazraa, Beirut, Lebanon
| | - Emanuela Szpotowicz
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Melitta Schachner
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, PR China.
| | - Igor Jakovcevski
- Center for Molecular Neurobiology Hamburg, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; Experimental Neurophysiology, University Hospital Cologne, Joseph-Stelzmann-Str. 9, 50931 Köln, Germany; German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| |
Collapse
|
45
|
Kruspe M, Thieme H, Guntinas-Lichius O, Irintchev A. Motoneuron regeneration accuracy and recovery of gait after femoral nerve injuries in rats. Neuroscience 2014; 280:73-87. [DOI: 10.1016/j.neuroscience.2014.08.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/27/2022]
|
46
|
Podestá MF, Yam P, Codagnone MG, Uccelli NA, Colman D, Reinés A. Distinctive PSA-NCAM and NCAM hallmarks in glutamate-induced dendritic atrophy and synaptic disassembly. PLoS One 2014; 9:e108921. [PMID: 25279838 PMCID: PMC4184824 DOI: 10.1371/journal.pone.0108921] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 09/05/2014] [Indexed: 12/13/2022] Open
Abstract
Dendritic and synapse remodeling are forms of structural plasticity that play a critical role in normal hippocampal function. Neural cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) participate in neurite outgrowth and synapse formation and plasticity. However, it remains unclear whether they contribute to dendritic retraction and synaptic disassembly. Cultured hippocampal neurons exposed to glutamate (5 µM) showed a reduced MAP-2 (+) area in the absence of neuronal death 24 h after the insult. Concomitantly, synapse loss, revealed by decreased synaptophysin and post-synaptic density-95 cluster number and area, together with changes in NCAM and PSA-NCAM levels were found. Dendritic atrophy and PSA-NCAM reduction proved NMDA-receptor dependent. Live-imaging experiments evidenced dendritic atrophy 4 h after the insult; this effect was preceded by smaller NCAM clusters (1 h) and decreased surface and total PSA-NCAM levels (3 h). Simultaneously, total NCAM cluster number and area remained unchanged. The subsequent synapse disassembly (6 h) was accompanied by reductions in total NCAM cluster number and area. A PSA mimetic peptide prevented both the dendritic atrophy and the subsequent synaptic changes (6 h) but had no effect on the earliest synaptic remodeling (3 h). Thus, NCAM-synaptic reorganization and PSA-NCAM level decrease precede glutamate-induced dendritic atrophy, whereas the NCAM level reduction is a delayed event related to synapse loss. Consequently, distinctive stages in PSA-NCAM/NCAM balance seem to accompany glutamate-induced dendritic atrophy and synapse loss.
Collapse
Affiliation(s)
- María Fernanda Podestá
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia Yam
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Martín Gabriel Codagnone
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Nonthué Alejandra Uccelli
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - David Colman
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Analía Reinés
- Instituto de Investigaciones Farmacológicas (ININFA, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN, CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
47
|
Pan HC, Shen YQ, Loers G, Jakovcevski I, Schachner M. Tegaserod, a small compound mimetic of polysialic acid, promotes functional recovery after spinal cord injury in mice. Neuroscience 2014; 277:356-66. [PMID: 25014876 DOI: 10.1016/j.neuroscience.2014.06.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/08/2014] [Accepted: 06/26/2014] [Indexed: 02/05/2023]
Abstract
In a previous study, we have shown that the small organic compound tegaserod, a drug approved for clinical application in an unrelated condition, is a mimic of the regeneration-beneficial glycan polysialic acid (PSA) in a mouse model of femoral nerve injury. Several independent observations have shown positive effects of PSA and its mimetic peptides in different paradigms of injury of the central and peripheral mammalian nervous systems. Since small organic compounds generally have advantages over metabolically rapidly degraded glycans and the proteolytically vulnerable mimetic peptides, a screen for a small PSA mimetic compound was successfully carried out, and the identified molecule proved to be beneficial in neurite outgrowth in vitro, independent of its originally described function as a 5-HT4 receptor agonist. In the present study, a mouse spinal cord compression device was used to elicit severe compression injury. We show that tegaserod promotes hindlimb motor function at 6 weeks after spinal cord injury compared to the control group receiving vehicle only. Immunohistology of the spinal cord rostral and caudal to the lesion site showed increased numbers of neurons, and a reduced area and intensity of glial fibrillary acidic protein immunoreactivity. Quantification of regrowth/sprouting of axons immunoreactive for tyrosine hydroxylase and serotonin showed increased axonal density rostral and caudal to the injury site in the ventral horns of mice treated with tegaserod. The combined observations suggest that tegaserod has the potential for treatment of spinal cord injuries in higher vertebrates.
Collapse
Affiliation(s)
- H-C Pan
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Y-Q Shen
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China; Jiangnan University Medical School, Wuxi, Jiangsu 214122, China
| | - G Loers
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg D-20246, Germany
| | - I Jakovcevski
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg D-20246, Germany
| | - M Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
48
|
Lutz D, Wolters-Eisfeld G, Schachner M, Kleene R. Cathepsin E generates a sumoylated intracellular fragment of the cell adhesion molecule L1 to promote neuronal and Schwann cell migration as well as myelination. J Neurochem 2014; 128:713-24. [DOI: 10.1111/jnc.12473] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/11/2013] [Accepted: 09/27/2013] [Indexed: 02/05/2023]
Affiliation(s)
- David Lutz
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Gerrit Wolters-Eisfeld
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience; Rutgers University; Piscataway New Jersey USA
- Center for Neuroscience; Shantou University Medical College; Shantou China
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| |
Collapse
|
49
|
Lo KWH, Jiang T, Gagnon KA, Nelson C, Laurencin CT. Small-molecule based musculoskeletal regenerative engineering. Trends Biotechnol 2014; 32:74-81. [PMID: 24405851 DOI: 10.1016/j.tibtech.2013.12.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 01/13/2023]
Abstract
Clinicians and scientists working in the field of regenerative engineering are actively investigating a wide range of methods to promote musculoskeletal tissue regeneration. Small-molecule-mediated tissue regeneration is emerging as a promising strategy for regenerating various musculoskeletal tissues and a large number of small-molecule compounds have been recently discovered as potential bioactive molecules for musculoskeletal tissue repair and regeneration. In this review, we summarize the recent literature encompassing the past 4 years in the area of small bioactive molecules for promoting repair and regeneration of various musculoskeletal tissues including bone, muscle, cartilage, tendon, and nerve.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA.
| | - Tao Jiang
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | - Keith A Gagnon
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | - Clarke Nelson
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Chemical, Materials and Biomolecular Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA.
| |
Collapse
|
50
|
Guseva D, Loers G, Schachner M. Function-triggering antibodies to the adhesion molecule L1 enhance recovery after injury of the adult mouse femoral nerve. PLoS One 2014; 9:e112984. [PMID: 25393007 PMCID: PMC4231121 DOI: 10.1371/journal.pone.0112984] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/19/2014] [Indexed: 02/05/2023] Open
Abstract
L1 is among the few adhesion molecules that favors repair after trauma in the adult central nervous system of vertebrates by promoting neuritogenesis and neuronal survival, among other beneficial features. In the peripheral nervous system, L1 is up-regulated in Schwann cells and regrowing axons after nerve damage, but the functional consequences of this expression remain unclear. Our previous study of L1-deficient mice in a femoral nerve injury model showed an unexpected improved functional recovery, attenuated motoneuronal cell death, and enhanced Schwann cell proliferation, being attributed to the persistent synthesis of neurotrophic factors. On the other hand, transgenic mice over-expressing L1 in neurons led to improved remyelination, but not improved functional recovery. The present study was undertaken to investigate whether the monoclonal L1 antibody 557 that triggers beneficial L1 functions in vitro would trigger these also in femoral nerve repair. We analyzed femoral nerve regeneration in C57BL/6J mice that received this antibody in a hydrogel filled conduit connecting the cut and sutured nerve before its bifurcation, leading to short-term release of antibody by diffusion. Video-based quantitative analysis of motor functions showed improved recovery when compared to mice treated with conduits containing PBS in the hydrogel scaffold, as a vehicle control. This improved recovery was associated with attenuated motoneuron loss, remyelination and improved precision of preferential motor reinnervation. We suggest that function-triggering L1 antibodies applied to the lesion site at the time of injury over a limited time period will not only be beneficial in peripheral, but also central nervous system regeneration.
Collapse
Affiliation(s)
- Daria Guseva
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|