1
|
Jiang M, Chen M, Liu N. Interactions between Schwann cell and extracellular matrix in peripheral nerve regeneration. Front Neurol 2024; 15:1372168. [PMID: 38651098 PMCID: PMC11034552 DOI: 10.3389/fneur.2024.1372168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/27/2024] [Indexed: 04/25/2024] Open
Abstract
Peripheral nerve injuries, caused by various reasons, often lead to severe sensory, motor, and autonomic dysfunction or permanent disability, posing a challenging problem in regenerative medicine. Autologous nerve transplantation has been the gold standard in traditional treatments but faces numerous limitations and risk factors, such as donor area denervation, increased surgical complications, and diameter or nerve bundle mismatches. The extracellular matrix (ECM) is a complex molecular network synthesized and released into the extracellular space by cells residing in tissues or organs. Its main components include collagen, proteoglycans/glycosaminoglycans, elastin, laminin, fibronectin, etc., providing structural and biochemical support to surrounding cells, crucial for cell survival and growth. Schwann cells, as the primary glial cells in the peripheral nervous system, play various important roles. Schwann cell transplantation is considered the gold standard in cell therapy for peripheral nerve injuries, making ECM derived from Schwann cells one of the most suitable biomaterials for peripheral nerve repair. To better understand the mechanisms of Schwann cells and the ECM in peripheral nerve regeneration and their optimal application, this review provides an overview of their roles in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Maorong Jiang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Muyang Chen
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Nana Liu
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Cho Y, Choi Y, Seong H. Nanoscale surface coatings and topographies for neural interfaces. Acta Biomater 2024; 175:55-75. [PMID: 38141934 DOI: 10.1016/j.actbio.2023.12.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
With the lack of minimally invasive tools for probing neuronal systems across spatiotemporal scales, understanding the working mechanism of the nervous system and limited assessments available are imperative to prevent or treat neurological disorders. In particular, nanoengineered neural interfaces can provide a solution to this technological barrier. This review covers recent surface engineering approaches, including nanoscale surface coatings, and a range of topographies from the microscale to the nanoscale, primarily focusing on neural-interfaced biosystems. Specifically, the immobilization of bioactive molecules to fertilize the neural cell lineage, topographical engineering to induce mechanotransduction in neural cells, and enhanced cell-chip coupling using three-dimensional structured surfaces are highlighted. Advances in neural interface design will help us understand the nervous system, thereby achieving the effective treatments for neurological disorders. STATEMENT OF SIGNIFICANCE: • This review focuses on designing bioactive neural interface with a nanoscale chemical modification and topographical engineering at multiscale perspective. • Versatile nanoscale surface coatings and topographies for neural interface are summarized. • Recent advances in bioactive materials applicable for neural cell culture, electrophysiological sensing, and neural implants are reviewed.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yunyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyejeong Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea.
| |
Collapse
|
3
|
Zhang G, Yang F, Li J, Chen S, Kong Y, Mo C, Leng X, Liu Y, Xu Y, Wang Y. A quinazoline derivative suppresses B cell hyper-activation and ameliorates the severity of systemic lupus erythematosus in mice. Front Pharmacol 2023; 14:1159075. [PMID: 37256224 PMCID: PMC10225574 DOI: 10.3389/fphar.2023.1159075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Background: Aberrant autoreactive B cell responses contribute to the pathogenesis of systemic lupus erythematosus (SLE). Currently, there is no safe and effective drug for intervention of SLE. Quinazoline derivative (N4-(4-phenoxyphenethyl)quinazoline-4,6-diamine, QNZ) is a NF-κB inhibitor and has potent anti-inflammatory activity. However, it is unclear whether QNZ treatment can modulate B cell activation and SLE severity. Methods: Splenic CD19+ B cells were treated with QNZ (2, 10, or 50 nM) or paeoniflorin (200 μM, a positive control), and their activation and antigen presentation function-related molecule expression were examined by flow cytometry. MRL/lpr lupus-prone mice were randomized and treated intraperitoneally with vehicle alone, 0.2 mg/kg/d QNZ or 1 mg/kg/d FK-506 (tacrolimus, a positive control) for 8 weeks. Their body weights and clinical symptoms were measured and the frequency of different subsets of splenic and lymph node activated B cells were quantified by flow cytometry. The degrees of kidney inflammation and glycogen deposition were examined by hematoxylin and eosin (H&E) and PAS staining. The levels of serum autoantibodies and renal IgG, complement C3 deposition were examined by ELISA and immunofluorescence. Results: QNZ treatment significantly inhibited the activation and antigen presentation-related molecule expression of B cells in vitro. Similarly, treatment with QNZ significantly mitigated the SLE activity by reducing the frequency of activated B cells and plasma cells in MRL/lpr mice. Conclusion: QNZ treatment ameliorated the severity of SLE in MRL/lpr mice, which may be associated with inhibiting B cell activation, and plasma cell formation. QNZ may be an excellent candidate for the treatment of SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Gan Zhang
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Fan Yang
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Juan Li
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Shan Chen
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Yuhang Kong
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Chunfen Mo
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xiao Leng
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Yang Liu
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Ying Xu
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Yantang Wang
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| |
Collapse
|
4
|
Caplan IF, Hernandez-Morato I, Pitman MJ. Temporal expression of Laminin-111 in the developing rat larynx. Neurosci Lett 2022; 781:136658. [PMID: 35483501 PMCID: PMC9194551 DOI: 10.1016/j.neulet.2022.136658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/08/2022] [Accepted: 04/22/2022] [Indexed: 12/31/2022]
Abstract
Laminin-111 is a basement membrane protein that participates in motor innervation and reinnervation. During axonal pathfinding, laminin-111 interacts with netrin-1 (NTN1) and changes its attractant growth cone properties into repulsion. While previous models of recurrent laryngeal nerve (RLN) transection show increased Laminin-111 and NTN1 production after injury, developmental expression in the larynx has not been defined. This study investigates the expression of laminin-111 in laryngeal muscles during primary laryngeal innervation of Sprague Dawley rats. Adult larynges and embryos were sectioned for immunohistochemistry with βIII-Tubulin, laminin subunit α-1 (LAMA1), NTN1, and α-bungarotoxin. Sections were processed for single-molecule inexpensive RNA fluorescence in situ hybridization analysis of LAMA1 mRNA. LAMA1 expression increased in all intrinsic laryngeal muscles, except the medial thyroarytenoid (MTA), at E20.5. At E20.5 there was increased expression in the lateral thyroarytenoid (LTA) and posterior cricoarytenoid (PCA) compared to the MTA. NTN1 upregulation was limited to the LTA and lateral cricoarytenoid (LCA) at E16.5 without any increase in the MTA or PCA. LAMA1 and NTN1 expression did not strictly follow expected patterns relative to the known timing of innervation and does not appear to be acting similarly to its role following RLN injury. These differences between developmental and post-injury innervation provide targets for investigations of therapeutics after nerve injury.
Collapse
Affiliation(s)
- Ian F. Caplan
- Columbia University Irving Medical Center/New York Presbyterian, Department of Otolaryngology Head & Neck Surgery, New York, NY, USA
| | - Ignacio Hernandez-Morato
- Columbia University Irving Medical Center/New York Presbyterian, Department of Otolaryngology Head & Neck Surgery, New York, NY, USA,Corresponding author at: Columbia University Irving Medical Center, Otolaryngology-Head and Neck Surgery, 180 Fort Washington Avenue, Rm 860 8th Floor. Harkness Pavilion, New York, NY 10032, USA. (I. Hernandez-Morato)
| | - Michael J. Pitman
- Columbia University Irving Medical Center/New York Presbyterian, Department of Otolaryngology Head & Neck Surgery, New York, NY, USA,Columbia University Irving Medical Center/New York Presbyterian, The Center for Voice and Swallowing, Department of Otolaryngology Head & Neck Surgery, New York, NY, USA
| |
Collapse
|
5
|
Blokland K, Pouwels S, Schuliga M, Knight D, Burgess J. Regulation of cellular senescence by extracellular matrix during chronic fibrotic diseases. Clin Sci (Lond) 2020; 134:2681-2706. [PMID: 33084883 PMCID: PMC7578566 DOI: 10.1042/cs20190893] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) is a complex network of macromolecules surrounding cells providing structural support and stability to tissues. The understanding of the ECM and the diverse roles it plays in development, homoeostasis and injury have greatly advanced in the last three decades. The ECM is crucial for maintaining tissue homoeostasis but also many pathological conditions arise from aberrant matrix remodelling during ageing. Ageing is characterised as functional decline of tissue over time ultimately leading to tissue dysfunction, and is a risk factor in many diseases including cardiovascular disease, diabetes, cancer, dementia, glaucoma, chronic obstructive pulmonary disease (COPD) and fibrosis. ECM changes are recognised as a major driver of aberrant cell responses. Mesenchymal cells in aged tissue show signs of growth arrest and resistance to apoptosis, which are indicative of cellular senescence. It was recently postulated that cellular senescence contributes to the pathogenesis of chronic fibrotic diseases in the heart, kidney, liver and lung. Senescent cells negatively impact tissue regeneration while creating a pro-inflammatory environment as part of the senescence-associated secretory phenotype (SASP) favouring disease progression. In this review, we explore and summarise the current knowledge around how aberrant ECM potentially influences the senescent phenotype in chronic fibrotic diseases. Lastly, we will explore the possibility for interventions in the ECM-senescence regulatory pathways for therapeutic potential in chronic fibrotic diseases.
Collapse
Affiliation(s)
- Kaj E.C. Blokland
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, Sydney, NSW, Australia
| | - Simon D. Pouwels
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
- Department of Lung Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | - Michael Schuliga
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
| | - Darryl A. Knight
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, Sydney, NSW, Australia
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Providence Health Care Research Institute, Vancouver, BC, Canada
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
6
|
Wu R, Chen B, Jia X, Qiu Y, Liu M, Huang C, Feng J, Wu Q. Interleukin-1β influences functional regeneration following nerve injury in mice through nuclear factor-κB signaling pathway. Immunology 2019; 156:235-248. [PMID: 30418673 DOI: 10.1111/imm.13022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 10/10/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022] Open
Abstract
This study focuses on investigating the role of interleukin-1β (IL-1β) in functional regeneration following nerve injury in mice. A microarray-based mRNA profiling study was used to analyze the expression level of IL-1β in peripheral nerve regeneration. Quantitative real-time polymerase chain reaction and Western blot were applied to assess the IL-1β expressions of C57BL/6J-crush and C57BL/6J-crush+IL-1β mice at different post-injury time-points after the standard sciatic nerve crush injury. The outcomes of nerve regeneration were evaluated by behavioral tests. IL-1β was found to be up-regulated in peripheral nerve regeneration and significantly raised on the 3rd day and returned to normal levels on the 14th day after nerve injury. Compared with C57BL/6J-crush+IL-1β mice, the nerve regeneration of C57BL/6J-crush mice was worse after nerve crush injury. IL-1β increased mechanical sensitivity and stimulated amplitude. IL-1β could benefit the recovery of sciatic nerve crush injury by facilitating nerve regeneration.
Collapse
Affiliation(s)
- Ruoyu Wu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bi Chen
- Department of Orthopedic Surgery, Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiang Jia
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu Qiu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mengyu Liu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengsheng Huang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jie Feng
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qingkai Wu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
7
|
Li G, Zhou R, Zhao X, Liu R, Ye C. Correlation between the expression of IL‑18 and deep venous thrombosis. Int J Mol Med 2018; 42:883. [PMID: 29786104 PMCID: PMC6034920 DOI: 10.3892/ijmm.2018.3682] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to investigate the effect of the expression of interleukin (IL)‑18 and related markers on deep venous thrombosis (DVT) to examine their correlation. Sprague‑Dawley rats of different models were established and were randomly assigned into three groups. The expression of IL‑18, nuclear factor (NF)‑κB and von Willebrand factor (vWF) were detected in blood samples. The inferior vena cava (IVC) was ligated to establish the DVT model. Rat IL‑18 overexpression and inhibition vectors were constructed. The expression levels of IL‑18 and related markers in the venous wall were compared between the model group and the control group using reverse transcription‑quantitative polymerase chain reaction and western blot analyses. Following the culture of human umbilical vein endothelial cells (HUVECs), IL‑18 was added to the cells, following which the growth of the HUVECs, and changes in vWF and other endothelial functional markers were analyzed. The IVC model demonstrated complete thrombosis at 8 h and stable thrombosis at 24 h. At 24 h following model establishment, the expression levels of IL‑18, NF‑κB and vWF were high in the blood samples with the occurrence and development of thrombosis (P<0.05). The weight, length and weight/length ratio of thrombi in each model group showed significant differences from those in the control group (P<0.05) with the overexpression of IL‑18, and the expression levels of NF‑κB and vWF in venous tissues were altered with abnormal expression levels of IL‑18. IL‑18 damaged HUVECs and significantly increased viability in early‑stage apoptosis, promoted the upregulation of vWF and P‑selectin, and reduced tissue plasminogen activator. IL‑18 and the related markers were closely associated with the occurrence and development of DVT.
Collapse
Affiliation(s)
- Guangdi Li
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Rudan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xueling Zhao
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Riguang Liu
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Chuan Ye
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
8
|
Bridging the Gap: Engineered Porcine-derived Urinary Bladder Matrix Conduits as a Novel Scaffold for Peripheral Nerve Regeneration. Ann Plast Surg 2018; 78:S328-S334. [PMID: 28328634 DOI: 10.1097/sap.0000000000001042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE This study aims to compare engineered nerve conduits constructed from porcine-derived urinary bladder matrix (UBM) with the criterion-standard nerve autografts, for segmental loss peripheral nerve repairs. METHODS Forty-eight Sprague-Dawley rats were divided into 2 groups. All underwent a 10-mm sciatic nerve gap injury. This was repaired using either (1) reverse autograft-the 10-mm cut segment was oriented 180 degrees and used to coapt the proximal and distal stumps or (2) UBM conduit-the 10-mm nerve gap was bridged with UBM conduit. Behavior assessments such as sciatic function index and foot fault asymmetry scores were performed weekly. At 3- or 6-week time endpoints, the repaired nerves and bilateral gastrocnemius/soleus muscles were harvested from each animal. Nerves were evaluated using immunohistochemistry for motor and sensory axon staining and with diffusion tensor imaging. The net wet muscle weights were calculated to assess the degree of muscle atrophy. RESULTS The UBM group demonstrated significantly improved foot fault asymmetry scores at 2 and 4 weeks, whereas there was no difference in sciatic function index. The net muscle weights were similar between both groups. Motor axon counts proximal/inside/distal to the conduit/graft were similar between UBM conduits and reverse autografts, whereas sensory axon counts within and distal to the conduit were significantly higher than those of the autograft at 6 weeks. Sensory axonal regeneration seemed to be adherent to the inner surface of the UBM conduit, whereas it had a scattered appearance in autografts. Diffusion tensor imaging parameters between groups were similar. CONCLUSIONS Urinary bladder matrix conduits prove to be at least similar to nerve autografts for the repair of peripheral nerve injuries with a short gap. The matrix perhaps serves as a scaffold to augment sensory nerve growth. CLINICAL RELEVANCE In a clinical setting, UBM may eliminate the donor site morbidity and increased operative time associated with nerve autografting.
Collapse
|
9
|
Abstract
Background: Laminins are extracellular matrix proteins that participate in endoneurial tubule formation and are important in the regeneration of nerves after injury. They act as scaffolds to guide nerves to distal targets and play a key role in neurite outgrowth. Because there is evidence that laminin architecture affects nerve regeneration, we evaluated endoneurial tubules by examining the laminin structure in clinical samples from patients with nerve injuries. Methods: In a retrospective review of eight nerve injury cases, we evaluated nerve histology in relation to clinical history and injury type. The immunohistochemical delineation of the laminin structure in relationship with the neuroma type was performed. Results: Five cases of upper-trunk stretch injuries—four from childbirth injury and one from a motorcycle accident—and three cases of nerve laceration leading to neuroma formation were examined. In the upper-trunk stretch injuries, avulsed nerves demonstrated no neuroma formation with a linear laminin architecture and a regular Schwann cell arrangement, but increased fibrous tissue deposition. For neuromas-in-continuity after a stretch injury, laminin immunohistochemistry demonstrated a double-lumen laminin tubule, with encapsulation of the Schwann cells and axonal processes. Nerve laceration leading to stump neuroma formation had a similar double-lumen laminin tubule, but less severe fibrosis. Conclusions: In nerve injuries with regenerative capacity, endoneurial tubules become pathologically disorganized. A double-lumen endoneurial tubule of unclear significance develops. The consistency of this pattern potentially suggests a reproducible pathophysiologic process. Further exploration of this pathophysiologic healing may provide insight into the failure of programmed peripheral nerve regeneration after injury.
Collapse
|
10
|
Ryan AJ, Lackington WA, Hibbitts AJ, Matheson A, Alekseeva T, Stejskalova A, Roche P, O'Brien FJ. A Physicochemically Optimized and Neuroconductive Biphasic Nerve Guidance Conduit for Peripheral Nerve Repair. Adv Healthc Mater 2017; 6. [PMID: 28975768 DOI: 10.1002/adhm.201700954] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Indexed: 11/07/2022]
Abstract
Clinically available hollow nerve guidance conduits (NGCs) have had limited success in treating large peripheral nerve injuries. This study aims to develop a biphasic NGC combining a physicochemically optimized collagen outer conduit to bridge the transected nerve, and a neuroconductive hyaluronic acid-based luminal filler to support regeneration. The outer conduit is mechanically optimized by manipulating crosslinking and collagen density, allowing the engineering of a high wall permeability to mitigate the risk of neuroma formation, while also maintaining physiologically relevant stiffness and enzymatic degradation tuned to coincide with regeneration rates. Freeze-drying is used to seamlessly integrate the luminal filler into the conduit, creating a longitudinally aligned pore microarchitecture. The luminal stiffness is modulated to support Schwann cells, with laminin incorporation further enhancing bioactivity by improving cell attachment and metabolic activity. Additionally, this biphasic NGC is shown to support neurogenesis and gliogenesis of neural progenitor cells and axonal outgrowth from dorsal root ganglia. These findings highlight the paradigm that a successful NGC requires the concerted optimization of both a mechanical support phase capable of bridging a nerve defect and a neuroconductive phase with an architecture capable of supporting both Schwann cells and neurons in order to achieve functional regenerative outcome.
Collapse
Affiliation(s)
- Alan J. Ryan
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - William A. Lackington
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - Alan J. Hibbitts
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - Austyn Matheson
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - Tijna Alekseeva
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - Anna Stejskalova
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - Phoebe Roche
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group (TERG); Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Trinity College Dublin; Dublin Ireland
- Trinity Centre for Bioengineering (TCBE); Trinity College Dublin; Dublin Ireland
| |
Collapse
|
11
|
The neurotrophic effects of different human dental mesenchymal stem cells. Sci Rep 2017; 7:12605. [PMID: 28974767 PMCID: PMC5626751 DOI: 10.1038/s41598-017-12969-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 09/18/2017] [Indexed: 12/15/2022] Open
Abstract
The current gold standard treatment for peripheral nerve injury is nerve grafting but this has disadvantages such as donor site morbidity. New techniques focus on replacing these grafts with nerve conduits enhanced with growth factors and/or various cell types such as mesenchymal stem cells (MSCs). Dental-MSCs (D-MSCs) including stem cells obtained from apical papilla (SCAP), dental pulp stem cells (DPSC), and periodontal ligament stem cells (PDLSC) are potential sources of MSCs for nerve repair. Here we present the characterization of various D-MSCs from the same human donors for peripheral nerve regeneration. SCAP, DPSC and PDLSC expressed BDNF, GDNF, NGF, NTF3, ANGPT1 and VEGFA growth factor transcripts. Conditioned media from D-MSCs enhanced neurite outgrowth in an in vitro assay. Application of neutralizing antibodies showed that brain derived neurotrophic factor plays an important mechanistic role by which the D-MSCs stimulate neurite outgrowth. SCAP, DPSC and PDLSC were used to treat a 10 mm nerve gap defect in a rat sciatic nerve injury model. All the stem cell types significantly enhanced axon regeneration after two weeks and showed neuroprotective effects on the dorsal root ganglia neurons. Overall the results suggested SCAP to be the optimal dental stem cell type for peripheral nerve repair.
Collapse
|
12
|
Thompson MJ, Patel G, Isaacs J, McMurtry J, Richards N, Daner W. Introduction of neurosupportive cells into processed acellular nerve allografts results in greater number and more even distribution when injected compared to soaking techniques. Neurol Res 2017; 39:189-197. [PMID: 28112028 DOI: 10.1080/01616412.2017.1282336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Processing necessary to remove immunogenic components of nerve allograft renders it acellular. Seeding with supportive cells may improve axon regeneration. We aim to identify the method associated with implantation of the greatest volume and most even distribution of cells. METHODS Hypodermic needle injection was compared to soaking in solution under both normal and pressurized conditions after micropuncture of the allograft. Distribution within the allograft was measured using an in vitro model of fluorescent beads, as well as cultured Schwann cells. RESULTS Injection treatment resulted in larger volumes and a more uniform cross-sectional distribution of implanted cells. Beads and cells behaved similarly relative to the measured outcomes. CONCLUSIONS Injection instills more cells in a more uniform distribution. In vivo testing may evaluate whether these techniques vary relative to cell survival, cell migration, and clinical outcomes. Size- and concentration-matched fluorescent beads may represent a viable model for analyzing cell implantation.
Collapse
Affiliation(s)
- Matthew J Thompson
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - Gaurangkumar Patel
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - Jonathan Isaacs
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - John McMurtry
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - Nathan Richards
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - William Daner
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| |
Collapse
|
13
|
Stimulating the proliferation, migration and lamellipodia of Schwann cells using low-dose curcumin. Neuroscience 2016; 324:140-50. [PMID: 26955781 DOI: 10.1016/j.neuroscience.2016.02.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/23/2016] [Accepted: 02/29/2016] [Indexed: 11/24/2022]
Abstract
Transplantation of peripheral glia is being trialled for neural repair therapies, and identification of compounds that enhance the activity of glia is therefore of therapeutic interest. We have previously shown that curcumin potently stimulates the activity of olfactory glia. We have now examined the effect of curcumin on Schwann cell (SC) activities including proliferation, migration and the expression of protein markers. SCs were treated with control media and with different concentrations of curcumin (0.02-20 μM). Cell proliferation was determined by MTS assay and migration changes were determined by single live cell migration tracking. We found that small doses of curcumin (40 nM) dramatically increased the proliferation and migration in SCs within just one day. When compared with olfactory glia, curcumin stimulated SC proliferation more rapidly and at lower concentrations. Curcumin significantly increased the migration of SCs, and also increased the dynamic activity of lamellipodial waves which are essential for SC migration. Expression of the activated form of the MAP kinase p38 (p-p38) was significantly decreased in curcumin-treated SCs. These results show that curcumin's effects on SCs differ remarkably to its effects on olfactory glia, suggesting that subtypes of closely related glia can be differentially stimulated by curcumin. Overall these results demonstrate that the therapeutically beneficial activities of glia can be differentially enhanced by curcumin which could be used to improve outcomes of neural repair therapies.
Collapse
|
14
|
Geuna S, Raimondo S, Fregnan F, Haastert-Talini K, Grothe C. In vitromodels for peripheral nerve regeneration. Eur J Neurosci 2015; 43:287-96. [DOI: 10.1111/ejn.13054] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/03/2015] [Accepted: 08/20/2015] [Indexed: 01/10/2023]
Affiliation(s)
- S. Geuna
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute; University of Turin; Ospedale San Luigi, Regione Gonzole 10 10043 Orbassano Turin Italy
| | - S. Raimondo
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute; University of Turin; Ospedale San Luigi, Regione Gonzole 10 10043 Orbassano Turin Italy
| | - F. Fregnan
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute; University of Turin; Ospedale San Luigi, Regione Gonzole 10 10043 Orbassano Turin Italy
| | - K. Haastert-Talini
- Institute of Neuroanatomy; Hannover Medical School and Center for Systems Neuroscience (ZSN); Hannover Germany
| | - C. Grothe
- Institute of Neuroanatomy; Hannover Medical School and Center for Systems Neuroscience (ZSN); Hannover Germany
| |
Collapse
|
15
|
Askvig JM, Watt JA. The MAPK and PI3K pathways mediate CNTF-induced neuronal survival and process outgrowth in hypothalamic organotypic cultures. J Cell Commun Signal 2015; 9:217-31. [PMID: 25698661 PMCID: PMC4580676 DOI: 10.1007/s12079-015-0268-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/05/2015] [Indexed: 11/26/2022] Open
Abstract
While collateral sprouting has been shown to occur in a variety of neuronal populations, the factor or factors responsible for mediating the sprouting response remain largely un-defined. There is evidence indicating that ciliary neurotrophic factor (CNTF) may play an important role in promoting neuronal survival and process outgrowth in neuronal phenotypes tested to date. We previously demonstrated that the astrocytic Jak-STAT pathway is necessary to mediate CNTF-induced oxytocinergic (OT) neuronal survival; however, the mechanism (s) of CNTF-mediated process outgrowth remain unknown. Our working hypothesis is that CNTF mediates differential neuroprotective responses via different intracellular signal transduction pathways. In order to test this hypothesis, we utilized stationary hypothalamic organotypic cultures to assess the contribution of the MAPK-ERK and PI3-AKT pathways to OT neuron survival and process outgrowth. Our results demonstrate that the MAPK-ERK½ pathway mediates CNTF-induced neuronal survival. Moreover, we show that inhibition of the p38-, JNK-MAPK, and mTOR pathways prevents loss OT neurons following axotomy. We also provide quantitative evidence indicating that CNTF promotes process outgrowth of OT neurons via the PI3K-AKT pathway. Together, these data indicate that distinct intracellular signaling pathways mediate diverse neuroprotective processes in response to CNTF.
Collapse
Affiliation(s)
- Jason M Askvig
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA.
| | - John A Watt
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Room 1701 Stop 9037, 501 N Columbia Road, Grand Forks, ND, 58203, USA.
| |
Collapse
|
16
|
de Luca AC, Lacour SP, Raffoul W, di Summa PG. Extracellular matrix components in peripheral nerve repair: how to affect neural cellular response and nerve regeneration? Neural Regen Res 2015; 9:1943-8. [PMID: 25598773 PMCID: PMC4283273 DOI: 10.4103/1673-5374.145366] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2014] [Indexed: 01/09/2023] Open
Abstract
Peripheral nerve injury is a serious problem affecting significantly patients’ life. Autografts are the “gold standard” used to repair the injury gap, however, only 50% of patients fully recover from the trauma. Artificial conduits are a valid alternative to repairing peripheral nerve. They aim at confining the nerve environment throughout the regeneration process, and providing guidance to axon outgrowth. Biocompatible materials have been carefully designed to reduce inflammation and scar tissue formation, but modifications of the inner lumen are still required in order to optimise the scaffolds. Biomicking the native neural tissue with extracellular matrix fillers or coatings showed great promises in repairing longer gaps and extending cell survival. In addition, extracellular matrix molecules provide a platform to further bind growth factors that can be released in the system over time. Alternatively, conduit fillers can be used for cell transplantation at the injury site, reducing the lag time required for endogenous Schwann cells to proliferate and take part in the regeneration process. This review provides an overview on the importance of extracellular matrix molecules in peripheral nerve repair.
Collapse
Affiliation(s)
- Alba C de Luca
- EPFL, Centre for Neuroprosthetics, Laboratory for Soft Bioelectronic Interfaces, Station 17, 1015 Lausanne, Switzerland
| | - Stephanie P Lacour
- EPFL, Centre for Neuroprosthetics, Laboratory for Soft Bioelectronic Interfaces, Station 17, 1015 Lausanne, Switzerland
| | - Wassim Raffoul
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Pietro G di Summa
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
17
|
Singh P, Zheng XL. Dual regulation of myocardin expression by tumor necrosis factor-α in vascular smooth muscle cells. PLoS One 2014; 9:e112120. [PMID: 25384061 PMCID: PMC4226488 DOI: 10.1371/journal.pone.0112120] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/12/2014] [Indexed: 12/31/2022] Open
Abstract
De-differentiation of vascular smooth muscle cells (VSMCs) plays a critical role in the development of atherosclerosis, a chronic inflammatory disease involving various cytokines such as tumor necrosis factor-α (TNFα). Myocardin is a co-factor of serum response factor (SRF) and is considered to be the master regulator of VSMC differentiation. It binds to SRF and regulates the expression of contractile proteins in VSMCs. Myocardin is also known to inhibit VSMC proliferation by inhibiting the NF-κB pathway, whereas TNFα is known to activate the NF-κB pathway in VSMCs. NF-κB activation has also been shown to inhibit myocardin expression and smooth muscle contractile marker genes. However, it is not definitively known whether TNFα regulates the expression and activity of myocardin in VSMCs. The current study aimed to investigate the role of TNFα in regulating myocardin and VSMC function. Our studies showed that TNFα down-regulated myocardin expression and activity in cultured VSMCs by activating the NF-κB pathway, resulting in decreased VSMC contractility and increased VSMC proliferation. Surprisingly, we also found that TNFα prevented myocardin mRNA degradation, and resulted in a further significant increase in myocardin expression and activity in differentiated VSMCs. Both the NF-κB and p44/42 MAPK pathways were involved in TNFα regulation of myocardin, which further increased the contractility of VSMCs. These differential effects of TNFα on myocardin seemingly depended on whether VSMCs were in a differentiated or de-differentiated state. Taken together, our results demonstrate that TNFα differentially regulates myocardin expression and activity, which may play a key role in regulating VSMC functions.
Collapse
Affiliation(s)
- Pavneet Singh
- Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xi-Long Zheng
- Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
18
|
Lin CY, Li LT, Su WT. Three dimensional chitosan scaffolds influence the extra cellular matrix expression in Schwann cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 42:474-8. [DOI: 10.1016/j.msec.2014.05.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/30/2014] [Accepted: 05/30/2014] [Indexed: 01/09/2023]
|
19
|
Shakhbazau A, Archibald SJ, Shcharbin D, Bryszewska M, Midha R. Aligned collagen-GAG matrix as a 3D substrate for Schwann cell migration and dendrimer-based gene delivery. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:1979-1989. [PMID: 24801062 DOI: 10.1007/s10856-014-5224-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/21/2014] [Indexed: 06/03/2023]
Abstract
The development of artificial off-the-shelf conduits that facilitate effective nerve regeneration and recovery after repair of traumatic nerve injury gaps is of fundamental importance. Collagen-glycosaminoglycan (GAG) matrix mimicking Schwann cell (SC) basal lamina has been proposed as a suitable and biologically rational substrate for nerve regeneration. In the present study, we have focused on the permissiveness of this matrix type for SC migration and repopulation, as these events play an essential role in nerve remodeling. We have also demonstrated that SCs cultured within collagen-GAG matrix are compatible with non-viral dendrimer-based gene delivery, that may allow conditioning of matrix-embedded cells for future gene therapy applications.
Collapse
Affiliation(s)
- Antos Shakhbazau
- Department of Clinical Neuroscience, Faculty of Medicine, University of Calgary, HMRB 109-3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada,
| | | | | | | | | |
Collapse
|
20
|
VEGF-A Promotes Both Pro-angiogenic and Neurotrophic Capacities for Nerve Recovery After Compressive Neuropathy in Rats. Mol Neurobiol 2014; 51:240-51. [DOI: 10.1007/s12035-014-8754-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 05/15/2014] [Indexed: 12/19/2022]
|
21
|
Tzekova N, Heinen A, Küry P. Molecules involved in the crosstalk between immune- and peripheral nerve Schwann cells. J Clin Immunol 2014; 34 Suppl 1:S86-104. [PMID: 24740512 DOI: 10.1007/s10875-014-0015-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
Abstract
Schwann cells are the myelinating glial cells of the peripheral nervous system and establish myelin sheaths on large caliber axons in order to accelerate their electrical signal propagation. Apart from this well described function, these cells revealed to exhibit a high degree of differentiation plasticity as they were shown to re- and dedifferentiate upon injury and disease as well as to actively participate in regenerative- and inflammatory processes. This review focuses on the crosstalk between glial- and immune cells observed in many peripheral nerve pathologies and summarizes functional evidences of molecules, regulators and factors involved in this process. We summarize data on Schwann cell's role presenting antigens, on interactions with the complement system, on Schwann cell surface molecules/receptors and on secreted factors involved in immune cell interactions or para-/autocrine signaling events, thus strengthening the view for a broader (patho) physiological role of this cell lineage.
Collapse
Affiliation(s)
- Nevena Tzekova
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, D-40225, Düsseldorf, Germany
| | | | | |
Collapse
|
22
|
Lu XM, Shu YH, Qiu CH, Chen KT, Wang YT. Protective effects and anti-apoptotic role of nerve growth factor on spinal cord neurons in sciatic nerve-injured rats. Neurol Res 2014; 36:814-23. [PMID: 24620979 DOI: 10.1179/1743132814y.0000000333] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The purpose of this study is to demonstrate a dependence of spinal cord motoneurons on the communication with their targets, sciatic nerves, and investigate whether the effects of nerve growth factor (NGF) on the spinal cord neuron apoptosis and surviving through the regulation of nuclear factor-kappa B (NF-kappaB) in Schwann cells (SCs) in sciatic nerve injured rats. METHODS Ninety healthy adult Sprague-Dawley rats were divided randomly into normal control group, crushing group, and NGF-intervened group. When sciatic nerve crushed 1, 3, 7, 14, and 21 days, the expression of NF-kappaB in SCs and the apoptosis regulator Bcl-2 and Caspase-3 in spinal cord were examined by immunohistochemistry staining, Western blot analysis, and immunofluorescence double-labeling method, the motor neuron apoptosis were investigated by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL), and the surviving neurons were tested by toluidine blue (Nissl) staining, respectively. All the data were further analyzed with SPSS10·0 application software. RESULTS The level of the expression of NF-kappaB in crushing group enhanced at 1 day after crushing, reached peak at 3 days, and reduced at least until 21 days, which was markedly higher than that in the normal control group. The expression of NF-kappaB in NGF-intervened group showed the same changes, reached peak at 7 days, and reduced until 21 days. However, when compared with crushing group, the expression of NF-kappaB in NGF-intervened group was down-regulated significantly until 3 days after injury, and up-regulated obviously with time going on. The same trend was observed in the time course on motor neuron apoptosis in crushing group and NGF-intervened group after sciatic nerves injury, while the reversing change was found in the surviving neurons. Moreover, the kinetics of Bcl-2 expression in spinal cord was consistent with that of NF-kappaB, while reversing with that of Caspase-3. CONCLUSION The findings revealed that NGF may play a pivotal role of anti-apoptosis in spinal cord neurons through retrograde transport of NF-kappaB in SCs following sciatic nerve injury in rats.
Collapse
|
23
|
Widgerow AD, Salibian AA, Kohan E, Sartiniferreira T, Afzel H, Tham T, Evans GRD. "Strategic sequences" in adipose-derived stem cell nerve regeneration. Microsurgery 2013; 34:324-30. [PMID: 24375471 DOI: 10.1002/micr.22219] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 12/08/2013] [Accepted: 12/12/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND Peripheral nerve injuries (PNI) are a major source of morbidity worldwide. The development of cellular regenerative therapies has the potential to improve outcomes of nerve injuries. However, an ideal therapy has yet to be found. The purpose of this study is to examine the current literature key points of regenerative techniques using human adipose-derived stem cells (hADSCs) for nerve regeneration and derive a comprehensive approach to hADSC therapy for PNI. METHODS A literature review was conducted using the electronic database PubMed to search for current experimental approaches to repairing PNI using hADSCs. Key search elements focused on specific components of nerve regeneration paradigms, including (1) support cells, (2) scaffolds, and (3) nerve conduits. RESULTS Strategic sequences were developed by optimizing the components of different experimental regenerative therapies. These sequences focus on priming hADSCs within a specialized growth medium, a hydrogel matrix base, and a collagen nerve conduit to achieve neuromodulatory nerve regeneration. hADSCs may exert their neuroregenerative influence through paracrine effects on surrounding Schwann cells in addition to physical interactions with injured tissue. CONCLUSIONS hADSCs may play a key role in nerve regeneration by acting primarily as support for local neurotrophic mediation and modulation of nerve growth rather than that of a primary neuronal differentiation agent.
Collapse
Affiliation(s)
- Alan D Widgerow
- Department of Aesthetic and Plastic Surgery, University of California, Irvine, CA
| | | | | | | | | | | | | |
Collapse
|
24
|
Purohit JS, Hu P, Chen G, Whelan J, Moustaid-Moussa N, Zhao L. Activation of nucleotide oligomerization domain containing protein 1 induces lipolysis through NF-κB and the lipolytic PKA activation in 3T3-L1 adipocytes. Biochem Cell Biol 2013; 91:428-434. [PMID: 24219284 DOI: 10.1139/bcb-2013-0049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Obesity is associated with chronic inflammation. Toll-like receptors (TLR) and NOD-like receptors (NLR) are two families of pattern recognition receptors that play important roles in the immune response and inflammation in adipocytes. Activation of TLR4 has been shown to stimulate lipolysis from adipose tissue or adipocytes. However, effects of activation of nucleotide-oligomerization domain containing protein 1 (NOD1), one of the prominent members of NLRs, on adipocyte lipolysis have not been studied. Here we report that NOD1 activation by the synthetic ligands (Tri-DAP and C12-iEDAP) stimulated lipolysis in 3T3-L1 adipocytes in a time- and dose-dependent manner. C12-iEDAP-induced lipolysis was attenuated with NOD1 siRNA knockdown, demonstrating the specificity of the effects. Moreover, inhibition of the protein kinase A (PKA)/hormone sensitive lipase (HSL) and NF-κB pathways by the pharmacological inhibitors attenuated the lipolytic effects of C12-iEDAP. Furthermore, we show NOD1 activation induced PKA activation independent of cAMP production and inhibition of NF-κB pathways attenuated phosphorylation of selected PKA lipolytic targets (phosphorylation of Perilipin Ser 517 and HSL Ser 563). Taken together, our results demonstrate a novel role of NOD1 activation, via NF-κB/PKA lipolytic activation, in inducing lipolysis in adipocytes and suggest that NOD1 activation may contribute to dyslipidemia in obesity.
Collapse
Affiliation(s)
- Jaanki S Purohit
- a Department of Nutrition, University of Tennessee, 1215 W. Cumberland Ave., Knoxville, TN, USA
| | | | | | | | | | | |
Collapse
|
25
|
Ogiwara T, Araki O, Morimura T, Tsunekawa K, Mori M, Murakami M. A novel mechanism for the inhibition of type 2 iodothyronine deiodinase by tumor necrosis factor α: involvement of proteasomal degradation. Endocr J 2013; 60:1035-45. [PMID: 23719846 DOI: 10.1507/endocrj.ej11-0144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Thyroxine (T₄) needs to be converted to 3,5,3'-triiodothyronine (T₃) by iodothyronine deiodinase to exert its biological activity. Recent studies revealed the presence of type 2 iodothyronine deiodinase (D2) in human thyroid tissue, human skeletal muscle and other tissues, suggesting that D2 is involved in maintaining plasma T₃ level in human. Tumor necrosis factor α (TNFα) is an inflammatory cytokine of which production is elevated in patients with nonthyroidal illness. Although several lines of evidence suggest the causal role of TNFα in nonthyroidal illness, detailed nature of the effect of TNFα on D2 remains unclear. In the present study, we identified D2 activity and D2 mRNA in TCO-1 cells, which were derived from human anaplastic thyroid carcinoma, and studied the mechanisms involved in the regulation of D2 expression by TNFα. The characteristics of the deiodinating activity in TCO-1 cells were compatible with those of D2 and Northern analysis demonstrated that D2 mRNA was expressed in TCO-1cells. D2 activity and D2 mRNA expression were rapidly increased by dibutyryl cAMP ((Bu)₂cAMP). TNFα showed an inhibitory effect on (Bu)₂cAMP-stimulated D2 activity in spite of little effect on (Bu)₂cAMP-stimulated D2 mRNA expression. MG132, a proteasome inhibitor abolished TNFα suppression of D2 activity whereas BAY11-7082 or 6-amino-4-(4-phenoxyphenylethylamino) quinazoline, inhibitors of nuclear factor-κB (NF-κB) failed to attenuate the effect of TNFα on D2 activity. These data suggest that a posttranslational mechanism through proteasomal degradation but not NF-κB activation is involved in the suppression of D2 by TNFα.
Collapse
Affiliation(s)
- Takayuki Ogiwara
- Department of Clinical Laboratory Medicine, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Lau TT, Wang DA. Bioresponsive hydrogel scaffolding systems for 3D constructions in tissue engineering and regenerative medicine. Nanomedicine (Lond) 2013; 8:655-68. [DOI: 10.2217/nnm.13.32] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Among the diversity of scaffolding systems available, hydrogel remains a popular choice for tissue engineering applications. The current state-of-the-art bioresponsive hydrogels demand intricate designs in pursuit of acquiring desired timely responses, such as controlled release of biological factors, changes in mechanical properties and scaffold degradation, at the same rate as the natural extracellular matrix. In this review, a variety of bioresponsive hydrogels are discussed; in particular, bioactive and biodegradable hydrogels that facilitate cellular development and tissue morphogenesis are highlighted. Bioactive hydrogels are designed to deliver biomolecules such as cell-adhesive moieties and instructive ligands at close proximity to the cell for better uptake or exposure. Biodegradable hydrogels provide transient scaffolding support for therapeutic cell settlement while gradually degrading in response to physical or enzymatic stimuli. In addition, biomechanical stimuli from hydrogels can induce mutual constructive responses on cells and, hence, will also be covered in this review.
Collapse
Affiliation(s)
- Ting Ting Lau
- Division of Bioengineering, School of Chemical & Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, N1.3-B2-13, 637457, Singapore
| | - Dong-An Wang
- Division of Bioengineering, School of Chemical & Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, N1.3-B2-13, 637457, Singapore
| |
Collapse
|
27
|
Paviolo C, Haycock JW, Yong J, Yu A, Stoddart PR, McArthur SL. Laser exposure of gold nanorods can increase neuronal cell outgrowth. Biotechnol Bioeng 2013; 110:2277-91. [PMID: 23456616 DOI: 10.1002/bit.24889] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/18/2013] [Accepted: 02/20/2013] [Indexed: 01/27/2023]
Abstract
The usage of gold nanoparticles (Au NPs) in biological applications has risen significantly over the last 10 years. With the wide variety of chemical and biological functionalization available and their distinctive optical properties, Au NPs are currently used in a range of biological applications including sensing, labeling, drug delivery, and imaging applications. Among the available particles, gold nanorods (Au NRs) are particularly useful because their optical absorption can be tuned across the visible to near infrared region. Here, we present a novel application of Au NRs associated with low power laser exposure of NG108-15 neuronal cells. When cells were irradiated with a 780 nm laser, the average number of neurons with neurites increased. A similar stimulatory effect was observed for cells that were cultured with poly-(4-styrenesulfonic acid)-coated and silica-coated Au NRs. Furthermore, when the NG108-15 cells were cultured with both bare and coated Au NRs and then irradiated with 1.2-7.5 W/cm(2) at 780 nm, they showed a neurite length increase of up to 25 µm versus control. To the best of our knowledge, this effect has never been reported before. While the pathways of the stimulation is not yet clear, the data presented here demonstrates that it is linked to the absorption of light by the Au NRs. These initial results open up new opportunities for peripheral nerve regeneration treatments and for novel approaches to addressing central nervous system axons following spinal cord injury.
Collapse
Affiliation(s)
- Chiara Paviolo
- Biotactical Engineering, Industrial Research Institute Swinburne (IRIS), Faculty of Engineering and Industrial Science, Swinburne University of Technology, PO Box 218, Hawthorn, Victoria 3122, Australia
| | | | | | | | | | | |
Collapse
|
28
|
di Summa PG, Kalbermatten DF, Raffoul W, Terenghi G, Kingham PJ. Extracellular matrix molecules enhance the neurotrophic effect of Schwann cell-like differentiated adipose-derived stem cells and increase cell survival under stress conditions. Tissue Eng Part A 2012; 19:368-79. [PMID: 22897220 DOI: 10.1089/ten.tea.2012.0124] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Since the first reports of induction of adipose-derived stem cells (ASC) into neuronal and glial cell phenotypes, expectations have increased regarding their use in tissue engineering applications for nerve repair. Cell adhesion to extracellular matrix (ECM) is a basic feature of survival, differentiation, and migration of Schwann cells (SC) during nerve regeneration, and fibronectin and laminin are two key molecules of this process. Interaction between ECM and SC-like differentiated ASC (dASC) could potentially improve the neurotrophic potential of the stem cells. We have investigated the effect of ECM molecules on SC-like dASC in terms of proliferation, adhesion, and cell viability. Fibronectin and laminin did not affect the proliferation of dASC when compared with cell adherent tissue culture plastic, but significantly improved viability and cell attachment when dASC were exposed to apoptotic conditions. To assess the influence of the ECM molecules on dASC neurotrophic activity, dASC were seeded onto ECM-coated culture inserts suspended above dorsal root ganglia (DRG) sensory neurons. Neurite outgrowth of DRG neurons was enhanced when dASC were seeded on fibronectin and laminin when compared with controls. When DRG neurons and dASC were in direct contact on the various surfaces there was significantly enhanced neurite outgrowth and coculture with laminin-conditioned dASC produced the longest neurites. Compared with primary SCs, dASC grown on laminin produced similar levels of neurite outgrowth in the culture insert experiments but neurite length was shorter in the direct contact groups. Anti β1 integrin blocking antibody could inhibit baseline and dASC evoked neurite elongation but had no effect on outgrowth mediated by laminin-conditioned dASC. ECM molecules had no effect on the levels of nerve growth factor and brain-derived neurotrophic factor secretion from dASC. The results of the study suggest that ECM molecules can significantly improve the potential of dASC for nerve regeneration.
Collapse
Affiliation(s)
- Pietro G di Summa
- Regenerative Biomedicine Group, Blond McIndoe Research Laboratories, The University of Manchester, Manchester, United Kingdom
| | | | | | | | | |
Collapse
|
29
|
Mammadov B, Mammadov R, Guler MO, Tekinay AB. Cooperative effect of heparan sulfate and laminin mimetic peptide nanofibers on the promotion of neurite outgrowth. Acta Biomater 2012; 8:2077-86. [PMID: 22342826 DOI: 10.1016/j.actbio.2012.02.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 02/01/2012] [Accepted: 02/06/2012] [Indexed: 01/01/2023]
Abstract
Extracellular matrix contains an abundant variety of signals that are received by cell surface receptors contributing to cell fate, via regulation of cellular activities such as proliferation, migration and differentiation. Cues from extracellular matrix can be used for the development of materials to direct cells into their desired fate. Neural extracellular matrix (ECM) is rich in axonal growth inducer proteins, and by mimicking these permissive elements in the cellular environment, neural differentiation as well as neurite outgrowth can be induced. In this paper, we used a synthetic peptide nanofiber system that can mimic not only the activity of laminin, an axonal growth-promoting constituent of the neural ECM, but also the activity of heparan sulfate proteoglycans in order to induce neuritogenesis. Heparan sulfate mimetic groups that were utilized in our system have an affinity to growth factors and induce the neuroregenerative effect of laminin mimetic peptide nanofibers. The self-assembled peptide nanofibers with heparan sulfate mimetic and laminin-derived epitopes significantly promoted neurite outgrowth by PC-12 cells. In addition, these scaffolds were even effective in the presence of chondroitin sulfate proteoglycans (CSPGs), which are the major inhibitory components of the central nervous system. In the presence of these nanofibers, cells could overcome CSPG inhibitory effect and extend neurites on peptide nanofiber scaffolds.
Collapse
|
30
|
Bell JHA, Haycock JW. Next generation nerve guides: materials, fabrication, growth factors, and cell delivery. TISSUE ENGINEERING PART B-REVIEWS 2011; 18:116-28. [PMID: 22010760 DOI: 10.1089/ten.teb.2011.0498] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nerve guides are increasingly being used surgically to repair acute peripheral nerve injuries. This is not only due to an increase in the number of commercially available devices, but also clinical acceptance. However, regeneration distance is typically limited to 20-25 mm, in part due to the basic tubular design. A number of experimental studies have shown improvements in nerve regeneration distance when conduits incorporate coatings, internal scaffolds, topographical cues, or the delivery of support cells. Current studies on designing nerve guides for maximizing nerve regeneration focus both on cell-containing and cell-free devices, the latter being clinically attractive as "off the shelf" products. Arguably better results are obtained when conduits are used in conjunction with support cells (e.g., Schwann cells or stem cells) that can improve regeneration distance and speed of repair, and provide informative experimental data on how Schwann and neuronal cells respond in regenerating injured nerves. In this review we discuss the range of current nerve guides commercially available and appraise experimental studies in the context of the future design of nerve guides for clinical use.
Collapse
Affiliation(s)
- Juliet H A Bell
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
| | | |
Collapse
|
31
|
Ding T, Lu WW, Zheng Y, Li ZY, Pan HB, Luo Z. Rapid repair of rat sciatic nerve injury using a nanosilver-embedded collagen scaffold coated with laminin and fibronectin. Regen Med 2011; 6:437-47. [PMID: 21749202 DOI: 10.2217/rme.11.39] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Scaffold with micro-channels has shown great promise in facilitating axonal regeneration after peripheral nerve injury. Significant research has focused on mimicking, in terms of composition and function, the ability of the basement membrane of Schwann cells to both promote and guide axonal regeneration. We aim to investigate the ability of a tissue-engineered scaffold with nanosilver and collagen to adsorb laminin and fibronectin, and the usefulness of this scaffold for repairing and regenerating a 10-mm peripheral nerve gap in rats. METHODS In this study, nanosilver-embedded collagen scaffolds were prepared and coated with laminin (LN) or LN plus fibronectin (FN). Scanning electron microscopy of the transverse and longitudinal sections of the scaffold revealed axially oriented microtubules ranging from 20 to 80 µm in diameter, and the internal surface of microtubules was found to be evenly coated with LN and FN. Energy dispersive spectrometry also confirmed an even distribution of nanosilver particles within the scaffold. To test its effectiveness in restoring neuronal connection, the scaffold was used in order to bridge 10 mm gaps in the severed sciatic nerve of rats. The rats were divided into an experimental group (receiving scaffold coated with LN and FN), a control group (receiving scaffold coated with LN only) and an autologous graft group. The functional recovery 40 days after surgery was examined by electrophysiology and sciatic nerve functional index (SFI) evaluation. FluoroGold™ (FG) retrograde tracing, toluidine blue staining and transmission electron microscopy were also used to examine the regenerated nerve fibers and to establish their myelination status. RESULTS The experimental group displayed partially restored nerve function. The recovery was comparable to the effect of autologous nerve graft and was better than that observed in the control group. A better functional recovery correlated with more FG-labeled neurons, higher density of toluidine blue stained nerve fibers and thicker myelin sheath. CONCLUSION Our results demonstrated that nanosilver-embedded collagen scaffolds with LN and FN coating is effective in aiding axonal regeneration, and recovery is comparable to the effect of an autologous nerve graft.
Collapse
Affiliation(s)
- Tan Ding
- Institute of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | |
Collapse
|
32
|
Interleukin 22 signaling promotes cell growth in mantle cell lymphoma. Transl Oncol 2011; 4:9-19. [PMID: 21286373 DOI: 10.1593/tlo.10172] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 10/05/2010] [Accepted: 10/07/2010] [Indexed: 12/28/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a specific type of aggressive B-cell non-Hodgkin lymphoma. We recently found that IL-22RA1, one of the two subunits of the interleukin 22 (IL-22) receptor, is expressed in MCL cell lines but not benign lymphocytes. In view of normal functions of IL-22 signaling, we hypothesized that the aberrant expression of IL-22RA1 may contribute to the deregulation of various cell signaling pathways, thereby promoting cell growth in MCL. In this study, we first demonstrated the expression of IL-22RA1 in all three MCL cell lines and eight frozen tumors examined using reverse transcription-polymerase chain reaction and Western blot analysis. In support of the concept that IL-22 signaling is biologically important in MCL, we found that MCL cells treated with recombinant IL-22 had a significant increase in cell growth that was associated with STAT3 activation. To investigate the mechanism underlying the aberrant expression of IL-22RA1, we analyzed the gene promoter of IL-22RA1, and we found multiple binding sites for NF-κB, a transcriptional factor strongly implicated in the pathogenesis of MCL. Pharmacologic inhibition of NF-κB resulted in a substantial reduction in the level of IL-22RA1 protein expression in MCL cells. To conclude, IL-22RA is aberrantly expressed in MCL, and we have provided evidence that IL-22 signaling contributes to the pathogenesis of MCL.
Collapse
|
33
|
Read DE, Gorman AM. Involvement of Akt in neurite outgrowth. Cell Mol Life Sci 2009; 66:2975-84. [PMID: 19504044 PMCID: PMC11115732 DOI: 10.1007/s00018-009-0057-8] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Revised: 05/19/2009] [Accepted: 05/20/2009] [Indexed: 02/06/2023]
Abstract
The regulation of neuronal differentiation and neurite outgrowth is essential during development of the nervous system and is crucial in developing therapies to promote axon regeneration after nerve injury or in neurodegenerative diseases. The serine/threonine kinase Akt has been well documented to promote neuronal survival. More recently Akt has also been revealed as key mediator of several aspects of neurite outgrowth, including elongation, branching and calibre. Downstream of Akt, several substrates have been identified that are likely to play key roles in Akt-mediated neurite outgrowth, such as glycogen synthase kinase 3beta, peripherin, mammalian target of rapamycin and delta-catenin. The physical interaction between Akt and Hsp27, another protein that has been linked with neurite outgrowth, may also be significant in the process of neurite outgrowth. This review will unite and discuss the research to date that has examined the functionality of Akt in neuronal differentiation during development and neurite outgrowth.
Collapse
Affiliation(s)
- Danielle E. Read
- Cell Death and Survival Group, Department of Biochemistry, School of Natural Sciences, National University of Ireland, Galway, Ireland
- National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Adrienne M. Gorman
- Cell Death and Survival Group, Department of Biochemistry, School of Natural Sciences, National University of Ireland, Galway, Ireland
- National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| |
Collapse
|
34
|
Conrad AH, Albrecht M, Pettit-Scott M, Conrad GW. Embryonic corneal Schwann cells express some Schwann cell marker mRNAs, but no mature Schwann cell marker proteins. Invest Ophthalmol Vis Sci 2009; 50:4173-84. [PMID: 19387082 PMCID: PMC2771572 DOI: 10.1167/iovs.08-3136] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Embryonic chick nerves encircle the cornea in pericorneal tissue until embryonic day (E)9, then penetrate the anterior corneal stroma, invade the epithelium, and branch over the corneal surface through E20. Adult corneal nerves, cut during transplantation or LASIK, never fully regenerate. Schwann cells (SCs) protect nerve fibers and augment nerve repair. This study evaluates SC differentiation in embryonic chick corneas. METHODS Fertile chicken eggs were incubated from E0 at 38 degrees C, 45% humidity. Dissected permeabilized corneas plus pericorneal tissue were immunostained for SC marker proteins. Other corneas were paraffin embedded, sectioned, and processed by in situ hybridization for corneal-, nerve-related, and SC marker gene expression. E9 to E20 corneas, dissected from pericorneal tissue, were assessed by real-time PCR (QPCR) for mRNA expression. RESULTS QPCR revealed unchanging low to moderate SLIT2/ROBO and NTN/UNC5 family, BACE1, and CADM3/CADM4 expressions, but high NEO1 expression. EGR2 and POU3F1 expressions never surpassed PAX3 expression. ITGNA6/ITGNB4 expressions increased 20-fold; ITGNB1 expression was high. SC marker S100 and MBP expressions increased; MAG, GFAP, and SCMP expressions were very low. Antibodies against the MPZ, MAG, S100, and SCMP proteins immunostained along pericorneal nerves, but not along corneal nerves. In the cornea, SLIT2 and SOX10 mRNAs were expressed in anterior stroma and epithelium, whereas PAX3, S100, MBP, and MPZL1 mRNAs were expressed only in corneal epithelium. CONCLUSIONS Embryonic chick corneas contain SCs, as defined by SOX10 and PAX3 transcription, which remain immature, at least in part because of stromal transcriptional and epithelial translational regulation of some SC marker gene expression.
Collapse
Affiliation(s)
- Abigail H Conrad
- Division of Biology, Kansas State University, Manhattan, Kansas 66506-4901, USA.
| | | | | | | |
Collapse
|
35
|
Peripheral neural cell sensitivity to mTHPC-mediated photodynamic therapy in a 3D in vitro model. Br J Cancer 2009; 101:658-65. [PMID: 19638975 PMCID: PMC2736832 DOI: 10.1038/sj.bjc.6605197] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: The effect of photodynamic therapy (PDT) on neural cells is important when tumours are within or adjacent to the nervous system. The purpose of this study was to investigate PDT using the photosensitiser, meta-tetrahydroxyphenyl chlorin (mTHPC), on rat neurons and satellite glia, compared with human adenocarcinoma cells (MCF-7). Methods: Fluorescence microscopy confirmed that mTHPC was incorporated into all three cell types. Sensitivity of cells exposed to mTHPC-PDT (0–10 μg ml–1) was determined in a novel 3-dimensional collagen gel culture system. Cell death was quantified using propidium iodide and cell types were distinguished using immunocytochemistry. In some cases, neuron survival was confirmed by measuring subsequent neurite growth in monolayer culture. Results: MCF-7s and satellite glia were significantly more sensitive to PDT than neurons. Importantly, 4 μg ml–1 mTHPC-PDT caused no significant neuron death compared with untreated controls but was sufficient to elicit substantial cell death in the other cell types. Initially, treatment reduced neurite length; neurons then extended neurites equivalent to those of untreated controls. The protocol was validated using hypericin (0–3 μg ml–1), which caused neuron death equivalent to other cell types. Conclusion: Neurons in culture can survive mTHPC-PDT under conditions sufficient to kill tumour cells and other nervous system cells.
Collapse
|
36
|
Maximum-entropy network analysis reveals a role for tumor necrosis factor in peripheral nerve development and function. Proc Natl Acad Sci U S A 2009; 106:12494-9. [PMID: 19597151 DOI: 10.1073/pnas.0902237106] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Gene regulatory interactions that shape developmental processes can often can be inferred from microarray analysis of gene expression, but most computational methods used require extensive datasets that can be difficult to generate. Here, we show that maximum-entropy network analysis allows extraction of genetic interactions from limited microarray datasets. Maximum-entropy networks indicated that the inflammatory cytokine TNF-alpha plays a pivotal role in Schwann cell-axon interactions, and these data suggested that TNF mediates its effects by orchestrating cytoplasmic movement and axon guidance. In vivo and in vitro experiments confirmed these predictions, showing that Schwann cells in TNF(-/-) peripheral sensory bundles fail to envelop axons efficiently, and that recombinant TNF can partially correct these defects. These data demonstrate the power of maximum-entropy network-based methods for analysis of microarray data, and they indicate that TNF-alpha plays a direct role in Schwann cell-axon communication.
Collapse
|
37
|
Sakimoto T, Kim TI, Ellenberg D, Fukai N, Jain S, Azar DT, Chang JH. Collagen XVIII and corneal reinnervation following keratectomy. FEBS Lett 2008; 582:3674-80. [PMID: 18840438 PMCID: PMC2617711 DOI: 10.1016/j.febslet.2008.09.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 09/14/2008] [Accepted: 09/25/2008] [Indexed: 10/21/2022]
Abstract
The significance of collagen XVIII in the regulation of corneal reinnervation remains largely unknown. We used whole-mount immunoconfocal microscopy to localize collagen XVIII to the nerve basement membrane of wild-type (WT) mouse corneas. Transmission electron microscopy showed corneal nerve disorganization in collagen XVIII knockout mice (col18a1(-/-)). Antibody 2H3-specific neurofilament colocalized with collagens XVIII and IV and laminin-2 in WT mouse corneas, but did not colocalize with collagen IV and laminin-2 in col18a1(-/-) mouse corneas. Following keratectomy, col18a1(-/-) mice displayed decreased corneal neurite extension compared to WT mice. Our data indicate that collagen XVIII may play an important role in corneal reinnervation after wounding.
Collapse
Affiliation(s)
- Tohru Sakimoto
- Massachusetts Eye and Ear Infirmary and Schepens Eye Research Institute, Harvard University, Boston, MA
| | - Tae-im Kim
- Massachusetts Eye and Ear Infirmary and Schepens Eye Research Institute, Harvard University, Boston, MA
| | - David Ellenberg
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL
| | - Naomi Fukai
- Harvard Medical School, Department of Oral and Developmental Biology, Harvard School of Dental Medicine, Boston, MA
| | - Sandeep Jain
- Massachusetts Eye and Ear Infirmary and Schepens Eye Research Institute, Harvard University, Boston, MA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL
| | - Dimitri T. Azar
- Massachusetts Eye and Ear Infirmary and Schepens Eye Research Institute, Harvard University, Boston, MA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL
| | - Jin-Hong Chang
- Massachusetts Eye and Ear Infirmary and Schepens Eye Research Institute, Harvard University, Boston, MA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|