1
|
Chang K, Wu JG, Ma TL, Hsu SH, Cho KS, Yu Z, Lennikov A, Ashok A, Rajagopalan A, Chen MH, Su WF, Utheim TP, Chen DF. Bioengineering strategy to promote CNS nerve growth and regeneration via chronic glutamate signaling. Acta Biomater 2024; 190:165-177. [PMID: 39427766 PMCID: PMC11614675 DOI: 10.1016/j.actbio.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 09/21/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Being part of the mature mammalian central nervous system, impairments of the retina and optic nerves caused by trauma or diseases often cannot be restored. Progressive degeneration of retinal ganglion cells (RGCs) in glaucoma and other optic neuropathies gradually leads to permanent vision loss, which currently has no cure. The purpose of this study is to develop a biocompatible scaffold to support RGC survival and guide axon growth, facilitating optic nerve repair and regeneration. We here report that electrical stimulation (ES) significantly promoted neurite outgrowth and elongation from primary RGCs, mediated through glutamate receptor signaling. To mimic prolonged glutamate stimulation and facilitate sustained nerve growth, we fabricated biocompatible poly-γ-benzyl-L-glutamate (PBG) scaffolds for controlled glutamate release. These PBG scaffolds supported RGC survival and robust long-distance nerve growth in both retinal explants and isolated RGC cultures. In contrast, control polycaprolactone (PCL) scaffolds with similar physical structures showed little benefits on RGC survival or nerve growth. Moreover, PBG scaffolds promoted the differentiation and neurite outgrowth from embryonic stem cell-derived RGC progenitors. The aligned PBG scaffold drove directed nerve elongation along the fiber alignment. Transplantation of PBG-coated biocompatible conduits induced robust optic nerve regeneration in adult mice following nerve transection. Together, the findings present the exciting possibility of driving optic nerve regeneration and RGC progenitor cell differentiation by imitating ES or glutamate signaling. PBG presents a permissive biomaterial in supporting robust and directed axon growth with promising clinical applications in the future. STATEMENT OF SIGNIFICANCE: We here reported compelling findings that demonstrate the potent regenerative effects of a bioengineered scaffold incorporating poly-γ-benzyl-L-glutamate (PBG) on the optic nerve. Retinal ganglion cell (RGC) axons, which form the optic nerve, are incapable of regenerating in adulthood, posing a significant hurdle in restoring vision for patients with optic nerve diseases or injuries. Built upon the finding that electrical stimulation promotes RGC axonal growth through glutamate signaling, we developed PBG scaffolds to provide sustained glutamate stimulation and showed their exceptional effects on driving directed axonal elongation in cultured RGCs and neural progenitors, as well as supporting robust optic nerve regeneration after transection in vivo. The findings hold great promise for reversing vision loss in patients with optic nerve conditions.
Collapse
Affiliation(s)
- Karen Chang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jhih-Guang Wu
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Tien-Li Ma
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hao Hsu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Zicheng Yu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Anton Lennikov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Ajay Ashok
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Aishwarya Rajagopalan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Biological Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Min-Huey Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Wei-Fang Su
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway; Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Department of Ophthalmology, Drammen Hospital, Drammen, Norway
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Mullagulova AI, Timechko EE, Solovyeva VV, Yakimov AM, Ibrahim A, Dmitrenko DD, Sufianov AA, Sufianova GZ, Rizvanov AA. Adeno-Associated Viral Vectors in the Treatment of Epilepsy. Int J Mol Sci 2024; 25:12081. [PMID: 39596149 PMCID: PMC11593886 DOI: 10.3390/ijms252212081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Epilepsy is a brain disorder characterized by a persistent predisposition to epileptic seizures. With various etiologies of epilepsy, a significant proportion of patients develop pharmacoresistance to antiepileptic drugs, which necessitates the search for new therapeutic methods, in particular, using gene therapy. This review discusses the use of adeno-associated viral (AAV) vectors in gene therapy for epilepsy, emphasizing their advantages, such as high efficiency of neuronal tissue transduction and low immunogenicity/cytotoxicity. AAV vectors provide the possibility of personalized therapy due to the diversity of serotypes and genomic constructs, which allows for increasing the specificity and effectiveness of treatment. Promising orientations include the modulation of the expression of neuropeptides, ion channels, transcription, and neurotrophic factors, as well as the use of antisense oligonucleotides to regulate seizure activity, which can reduce the severity of epileptic disorders. This review summarizes the current advances in the use of AAV vectors for the treatment of epilepsy of various etiologies, demonstrating the significant potential of AAV vectors for the development of personalized and more effective approaches to reducing seizure activity and improving patient prognosis.
Collapse
Affiliation(s)
- Aysilu I. Mullagulova
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
| | - Elena E. Timechko
- Department of Medical Genetics and Clinical Neurophysiology, Krasnoyarsk State Medical University, Partizana Zheleznyaka 1, Krasnoyarsk 660022, Russia; (E.E.T.); (A.M.Y.); (D.D.D.)
| | - Valeriya V. Solovyeva
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
| | - Alexey M. Yakimov
- Department of Medical Genetics and Clinical Neurophysiology, Krasnoyarsk State Medical University, Partizana Zheleznyaka 1, Krasnoyarsk 660022, Russia; (E.E.T.); (A.M.Y.); (D.D.D.)
| | - Ahmad Ibrahim
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
| | - Diana D. Dmitrenko
- Department of Medical Genetics and Clinical Neurophysiology, Krasnoyarsk State Medical University, Partizana Zheleznyaka 1, Krasnoyarsk 660022, Russia; (E.E.T.); (A.M.Y.); (D.D.D.)
| | - Albert A. Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119991, Russia;
- The Research and Educational Institute of Neurosurgery, Peoples’ Friendship University of Russia, Moscow 117198, Russia
| | - Galina Z. Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen 625023, Russia;
| | - Albert A. Rizvanov
- Institute for Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (A.I.M.); (V.V.S.); (A.I.)
- Division of Medical and Biological Sciences, Academy of Sciences of the Republic of Tatarstan, Kazan 420111, Russia
| |
Collapse
|
3
|
Zhou W, Rahman MSU, Sun C, Li S, Zhang N, Chen H, Han CC, Xu S, Liu Y. Perspectives on the Novel Multifunctional Nerve Guidance Conduits: From Specific Regenerative Procedures to Motor Function Rebuilding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307805. [PMID: 37750196 DOI: 10.1002/adma.202307805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Indexed: 09/27/2023]
Abstract
Peripheral nerve injury potentially destroys the quality of life by inducing functional movement disorders and sensory capacity loss, which results in severe disability and substantial psychological, social, and financial burdens. Autologous nerve grafting has been commonly used as treatment in the clinic; however, its rare donor availability limits its application. A series of artificial nerve guidance conduits (NGCs) with advanced architectures are also proposed to promote injured peripheral nerve regeneration, which is a complicated process from axon sprouting to targeted muscle reinnervation. Therefore, exploring the interactions between sophisticated NGC complexes and versatile cells during each process including axon sprouting, Schwann cell dedifferentiation, nerve myelination, and muscle reinnervation is necessary. This review highlights the contribution of functional NGCs and the influence of microscale biomaterial architecture on biological processes of nerve repair. Progressive NGCs with chemical molecule induction, heterogenous topographical morphology, electroactive, anisotropic assembly microstructure, and self-powered electroactive and magnetic-sensitive NGCs are also collected, and they are expected to be pioneering features in future multifunctional and effective NGCs.
Collapse
Affiliation(s)
- Weixian Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Nuozi Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Hao Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Charles C Han
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
4
|
Bianchini M, Micera S, Redolfi Riva E. Recent Advances in Polymeric Drug Delivery Systems for Peripheral Nerve Regeneration. Pharmaceutics 2023; 15:pharmaceutics15020640. [PMID: 36839962 PMCID: PMC9965241 DOI: 10.3390/pharmaceutics15020640] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
When a traumatic event causes complete denervation, muscle functional recovery is highly compromised. A possible solution to this issue is the implantation of a biodegradable polymeric tubular scaffold, providing a biomimetic environment to support the nerve regeneration process. However, in the case of consistent peripheral nerve damage, the regeneration capabilities are poor. Hence, a crucial challenge in this field is the development of biodegradable micro- nanostructured polymeric carriers for controlled and sustained release of molecules to enhance nerve regeneration. The aim of these systems is to favor the cellular processes that support nerve regeneration to increase the functional recovery outcome. Drug delivery systems (DDSs) are interesting solutions in the nerve regeneration framework, due to the possibility of specifically targeting the active principle within the site of interest, maximizing its therapeutical efficacy. The scope of this review is to highlight the recent advances regarding the study of biodegradable polymeric DDS for nerve regeneration and to discuss their potential to enhance regenerative performance in those clinical scenarios characterized by severe nerve damage.
Collapse
Affiliation(s)
- Marta Bianchini
- The BioRobotics Institute, Department of Excellence in Robotics and AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Silvestro Micera
- The BioRobotics Institute, Department of Excellence in Robotics and AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Translational Neuroengineering, Centre for Neuroprosthetics and Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1000 Lausanne, Switzerland
| | - Eugenio Redolfi Riva
- The BioRobotics Institute, Department of Excellence in Robotics and AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Correspondence:
| |
Collapse
|
5
|
Khotimchenko YS, Silachev DN, Katanaev VL. Marine Natural Products from the Russian Pacific as Sources of Drugs for Neurodegenerative Diseases. Mar Drugs 2022; 20:708. [PMID: 36421986 PMCID: PMC9697637 DOI: 10.3390/md20110708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 09/05/2023] Open
Abstract
Neurodegenerative diseases are growing to become one of humanity's biggest health problems, given the number of individuals affected by them. They cause enough mortalities and severe economic impact to rival cancers and infections. With the current diversity of pathophysiological mechanisms involved in neurodegenerative diseases, on the one hand, and scarcity of efficient prevention and treatment strategies, on the other, all possible sources for novel drug discovery must be employed. Marine pharmacology represents a relatively uncharted territory to seek promising compounds, despite the enormous chemodiversity it offers. The current work discusses one vast marine region-the Northwestern or Russian Pacific-as the treasure chest for marine-based drug discovery targeting neurodegenerative diseases. We overview the natural products of neurological properties already discovered from its waters and survey the existing molecular and cellular targets for pharmacological modulation of the disease. We further provide a general assessment of the drug discovery potential of the Russian Pacific in case of its systematic development to tackle neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuri S. Khotimchenko
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 8 ul. Sukhanova, 690950 Vladivostok, Russia
- A.V. Zhirmunsky National Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690950 Vladivostok, Russia
| | - Denis N. Silachev
- Department of Functional Biochemistry of Biopolymers, A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Vladimir L. Katanaev
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 8 ul. Sukhanova, 690950 Vladivostok, Russia
- Department of Cell Physiology and Metabolism, Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| |
Collapse
|
6
|
Guo Y, Wang S, Chao X, Li D, Wang Y, Guo Q, Chen T. Multi-omics studies reveal ameliorating effects of physical exercise on neurodegenerative diseases. Front Aging Neurosci 2022; 14:1026688. [PMID: 36389059 PMCID: PMC9659972 DOI: 10.3389/fnagi.2022.1026688] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/28/2022] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease, are heavy burdens to global health and economic development worldwide. Mounting evidence suggests that exercise, a type of non-invasive intervention, has a positive impact on the life quality of elderly with neurodegenerative diseases. X-omics are powerful tools for mapping global biochemical changes in disease and treatment. METHOD Three major databases were searched related to current studies in exercise intervention on neurodegenerative diseases using omics tools, including metabolomics, metagenomics, genomics, transcriptomics, and proteomics. RESULT We summarized the omics features and potential mechanisms associated with exercise and neurodegenerative diseases in the current studies. Three main mechanisms by which exercise affects neurodegenerative diseases were summed up, including adult neurogenesis, brain-derived neurotrophic factor (BDNF) signaling, and short-chain fatty acids (SCFAs) metabolism. CONCLUSION Overall, there is compelling evidence that exercise intervention is a feasible way of preventing the onset and alleviating the severity of neurodegenerative diseases. These studies highlight the importance of exercise as a complementary approach to the treatment and intervention of neurodegenerative diseases in addition to traditional treatments. More mechanisms on exercise interventions for neurodegenerative diseases, the specification of exercise prescriptions, and differentiated exercise programs should be explored so that they can actually be applied to the clinic.
Collapse
Affiliation(s)
- Yuhuai Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shouli Wang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaowen Chao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ding Li
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Tianlu Chen
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
7
|
Rao Z, Lin Z, Song P, Quan D, Bai Y. Biomaterial-Based Schwann Cell Transplantation and Schwann Cell-Derived Biomaterials for Nerve Regeneration. Front Cell Neurosci 2022; 16:926222. [PMID: 35836742 PMCID: PMC9273721 DOI: 10.3389/fncel.2022.926222] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Schwann cells (SCs) dominate the regenerative behaviors after peripheral nerve injury by supporting axonal regrowth and remyelination. Previous reports also demonstrated that the existence of SCs is beneficial for nerve regeneration after traumatic injuries in central nervous system. Therefore, the transplantation of SCs/SC-like cells serves as a feasible cell therapy to reconstruct the microenvironment and promote nerve functional recovery for both peripheral and central nerve injury repair. However, direct cell transplantation often leads to low efficacy, due to injection induced cell damage and rapid loss in the circulatory system. In recent years, biomaterials have received great attention as functional carriers for effective cell transplantation. To better mimic the extracellular matrix (ECM), many biodegradable materials have been engineered with compositional and/or topological cues to maintain the biological properties of the SCs/SCs-like cells. In addition, ECM components or factors secreted by SCs also actively contribute to nerve regeneration. Such cell-free transplantation approaches may provide great promise in clinical translation. In this review, we first present the current bio-scaffolds engineered for SC transplantation and their achievement in animal models and clinical applications. To this end, we focus on the physical and biological properties of different biomaterials and highlight how these properties affect the biological behaviors of the SCs/SC-like cells. Second, the SC-derived biomaterials are also reviewed and discussed. Finally, the relationship between SCs and functional biomaterials is summarized, and the trends of their future development are predicted toward clinical applications.
Collapse
Affiliation(s)
- Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zudong Lin
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Panpan Song
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Daping Quan
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Endurance Training and Exogenous Adenosine Infusion Mitigate Hippocampal Inflammation and Cell Death in a Rat Model of Cerebral Ischemia/Reperfusion Injury. ARCHIVES OF NEUROSCIENCE 2022. [DOI: 10.5812/ans.119236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Cerebral ischemia can cause irreversible structural and functional damages to the brain, especially to the hippocampus. Preconditioning with endurance training and endogenous adenosine infusion may reduce ischemia-associated damages. Objectives: This study aimed to evaluate the effect of preconditioning with endurance training and endogenous adenosine infusion on cell death in the hippocampal CA1 region following ischemia/reperfusion injuries in a rat model. Methods: Male Wistar rats were divided into five groups: (1) control (n = 8); (2) ischemia (n = 12); (3) endurance training + ischemia (n = 12); (4) adenosine infusion + ischemia (n = 12); and (5) endurance training + adenosine infusion + ischemia (n = 12). The rats in the training groups ran on a treadmill five days per week for eight weeks. In the adenosine infusion groups, the rats were injected 0.1 mg/mL/kg of adenosine intraperitoneally. Also, in the ischemic groups, both common carotid arteries were clamped for 45 minutes. Cresyl violet staining and real-time polymerase chain reaction (PCR) assay were used to evaluate cell death and cytokine gene expression, respectively. Results: Based on the present results, treatments, including endurance training + ischemia, adenosine infusion + ischemia, and endurance training + adenosine infusion + ischemia reduced the level of interleukin-6 (IL-6) and glutamate gene expression, respectively, compared to the group of ischemia only. In contrast, the expression of nerve growth factor (NGF) and adenosine receptor (A2A) genes increased by seven, four, and two folds in the endurance training + ischemia, adenosine infusion + ischemia, and endurance training + adenosine infusion + ischemia groups, respectively, compared to the group of ischemia only. Conclusions: Endurance training on a treadmill and exogenous adenosine infusion synergistically diminished cell death and reduced the expression of pro-inflammatory cytokines, while promoting the neurotrophic factor expression. When endurance training and adenosine infusion were used as stimulants before the induction of cerebral ischemia, they significantly reduced cell death.
Collapse
|
9
|
Xia J, Yu P, Zeng Z, Ma M, Yan X, Zhao J, Gong D, Zhang G, Wang J. Medium chain triglycerides improve lipid metabolism in obese rats by increasing the browning of adipose tissue through the sympathetic regulation. Food Funct 2022; 13:8068-8080. [DOI: 10.1039/d2fo00239f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study aimed to determine the mechanism of medium chain triglyceride (MCT) promoting the browning of adipose tissue. High fat diet was fed to the Sprague-Dawley rats to induce obesity,...
Collapse
|
10
|
ExplantAnalyzer: An advanced automated neurite outgrowth analysis evaluated by means of organotypic auditory neuron explant cultures. J Neurosci Methods 2021; 363:109341. [PMID: 34474047 DOI: 10.1016/j.jneumeth.2021.109341] [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: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neuronal outgrowth assays using organotypic explant cultures are commonly utilized to study neuroregenerative and -protective effects of drugs such as neurotrophins. While this approach offers higher organized tissue compared to single cell cultures and less experimental effort than in-vivo studies, quantitative evaluation of the neuronal network is often time consuming. Thus, we developed ExplantAnlayzer, a time-saving high-throughput evaluation method, yielding numerous metrics to objectively describe neuronal outgrowth. NEW METHOD Spiral ganglion explants were cultured in 24-well plates, mechanically fixed in a collagen matrix and immunolabeled against beta-III-tubulin. The explants were imaged using a fluorescent tile-scan microscope and resulting images were stitched. The evaluation was developed as an open-source MATLAB routine and involves several image processing steps, including adaptive thresholding. The neurite network was eventually converted to a graph to track neurites from their terminals back to the explant body. COMPARISON WITH EXISTING METHOD(S) We compared ExplantAnlayzer quantitatively and qualitatively to common existing methods, such as Sholl analyses and manual fiber tracing, using representative explant images. ExplantAnlayzer is able to achieve similar and as detailed results as manual tracing while decreasing manual interaction and required time dramatically. RESULTS After an initial setup phase, the explant images could be batch-processed altogether. Bright bundles as well as faint fibers were reliably detected. Several metrics describing the outgrowth morphology, including total outgrowth, neurite numbers and length estimations, as well as their growth directions, were computed. CONCLUSIONS ExplantAnalyzer is a time-saving and objective method for an in-depth evaluation of organotypic explant outgrowth.
Collapse
|
11
|
Leal-Galicia P, Chávez-Hernández ME, Mata F, Mata-Luévanos J, Rodríguez-Serrano LM, Tapia-de-Jesús A, Buenrostro-Jáuregui MH. Adult Neurogenesis: A Story Ranging from Controversial New Neurogenic Areas and Human Adult Neurogenesis to Molecular Regulation. Int J Mol Sci 2021; 22:11489. [PMID: 34768919 PMCID: PMC8584254 DOI: 10.3390/ijms222111489] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
The generation of new neurons in the adult brain is a currently accepted phenomenon. Over the past few decades, the subventricular zone and the hippocampal dentate gyrus have been described as the two main neurogenic niches. Neurogenic niches generate new neurons through an asymmetric division process involving several developmental steps. This process occurs throughout life in several species, including humans. These new neurons possess unique properties that contribute to the local circuitry. Despite several efforts, no other neurogenic zones have been observed in many years; the lack of observation is probably due to technical issues. However, in recent years, more brain niches have been described, once again breaking the current paradigms. Currently, a debate in the scientific community about new neurogenic areas of the brain, namely, human adult neurogenesis, is ongoing. Thus, several open questions regarding new neurogenic niches, as well as this phenomenon in adult humans, their functional relevance, and their mechanisms, remain to be answered. In this review, we discuss the literature and provide a compressive overview of the known neurogenic zones, traditional zones, and newly described zones. Additionally, we will review the regulatory roles of some molecular mechanisms, such as miRNAs, neurotrophic factors, and neurotrophins. We also join the debate on human adult neurogenesis, and we will identify similarities and differences in the literature and summarize the knowledge regarding these interesting topics.
Collapse
Affiliation(s)
- Perla Leal-Galicia
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - María Elena Chávez-Hernández
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Florencia Mata
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Jesús Mata-Luévanos
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Luis Miguel Rodríguez-Serrano
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
- Laboratorio de Neurobiología de la Alimentación, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Alejandro Tapia-de-Jesús
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Mario Humberto Buenrostro-Jáuregui
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| |
Collapse
|
12
|
Falo CP, Benitez R, Caro M, Morell M, Forte-Lago I, Hernandez-Cortes P, Sanchez-Gonzalez C, O’Valle F, Delgado M, Gonzalez-Rey E. The Neuropeptide Cortistatin Alleviates Neuropathic Pain in Experimental Models of Peripheral Nerve Injury. Pharmaceutics 2021; 13:pharmaceutics13070947. [PMID: 34202793 PMCID: PMC8309056 DOI: 10.3390/pharmaceutics13070947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain is one of the most severe forms of chronic pain caused by the direct injury of the somatosensory system. The current drugs for treating neuropathies have limited efficacies or show important side effects, and the development of analgesics with novel modes of action is critical. The identification of endogenous anti-nociceptive factors has emerged as an attractive strategy for designing new pharmacological approaches to treat neuropathic pain. Cortistatin is a neuropeptide with potent anti-inflammatory activity, recently identified as a natural analgesic peptide in several models of pain evoked by inflammatory conditions. Here, we investigated the potential analgesic effect of cortistatin in neuropathic pain using a variety of experimental models of peripheral nerve injury caused by chronic constriction or partial transection of the sciatic nerve or by diabetic neuropathy. We found that the peripheral and central injection of cortistatin ameliorated hyperalgesia and allodynia, two of the dominant clinical manifestations of chronic neuropathic pain. Cortistatin-induced analgesia was multitargeted, as it regulated the nerve damage-induced hypersensitization of primary nociceptors, inhibited neuroinflammatory responses, and enhanced the production of neurotrophic factors both at the peripheral and central levels. We also demonstrated the neuroregenerative/protective capacity of cortistatin in a model of severe peripheral nerve transection. Interestingly, the nociceptive system responded to nerve injury by secreting cortistatin, and a deficiency in cortistatin exacerbated the neuropathic pain responses and peripheral nerve dysfunction. Therefore, cortistatin-based therapies emerge as attractive alternatives for treating chronic neuropathic pain of different etiologies.
Collapse
Affiliation(s)
- Clara P. Falo
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
| | - Raquel Benitez
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
| | - Marta Caro
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
| | - Maria Morell
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
- Genyo Center for Genomics and Oncological Research, Parque Tecnologico de la Salud, 18016 Granada, Spain
| | - Irene Forte-Lago
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
| | - Pedro Hernandez-Cortes
- Department of Orthopedic Surgery, San Cecilio University Hospital, 18071 Granada, Spain;
| | - Clara Sanchez-Gonzalez
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
| | - Francisco O’Valle
- Department of Pathology, School of Medicine, IBIMER and IBS-Granada, Granada University, 18016 Granada, Spain;
| | - Mario Delgado
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
- Correspondence: (M.D.); (E.G.-R.)
| | - Elena Gonzalez-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra, IPBLN-CSIC, Parque Tecnologico de la Salud, 18016 Granada, Spain; (C.P.F.); (R.B.); (M.C.); (M.M.); (I.F.-L.); (C.S.-G.)
- Correspondence: (M.D.); (E.G.-R.)
| |
Collapse
|
13
|
Amiri A, Kashani MHG, Ghorbanian MT. Expression of neurotrophic factor genes by human adipose stem cells post-induction by deprenyl. Anat Cell Biol 2021; 54:74-82. [PMID: 33526752 PMCID: PMC8017458 DOI: 10.5115/acb.19.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 10/16/2020] [Accepted: 11/03/2020] [Indexed: 11/28/2022] Open
Abstract
Human adipose stem cells (hASCs) were introduced as appropriate candidate due to advantages like ease of isolation, in vitro expansion and lack of immune response. Deprenyl (Dep) was used to induce bone marrow stem cells into neuron-like cells. We investigated the Dep effect on neurotrophin genes expression in hASCs and their differentiation into neuron-like cells. The cells were isolated from small pieces of abdominal adipose tissue and subjected to flow cytometry to confirm purification. The osteogenic and adipogenic differentiation were identified. The proliferation rate and neurotrophin genes expression of treated cells were evaluated by MTT, TH immunostaining and RT-PCR. hASCs had positive response to CD44, CD73, CD90, CD105 markers and negative response to CD34 and CD45 markers and differentiated into adipocytes and osteocytes. Exposure to 10–7 M of Dep for 24 hours caused a significant increase of viable cells and BDNF, NTF-3 genes expression as compared to cultured cells in serum free medium and had no effect on the expression of NGF and GDNF genes. Based on our results, Dep is able to induce BDNF, NTF-3 and NTF-4 genes expression and neroun-like morphology in hASCs.
Collapse
Affiliation(s)
- Arezoo Amiri
- Department of Cellular and Molecular Biology, School of Biology, Damghan University, Damghan, Iran
| | | | | |
Collapse
|
14
|
Carvalho CR, Chang W, Silva‐Correia J, Reis RL, Oliveira JM, Kohn J. Engineering Silk Fibroin-Based Nerve Conduit with Neurotrophic Factors for Proximal Protection after Peripheral Nerve Injury. Adv Healthc Mater 2021; 10:e2000753. [PMID: 33169544 DOI: 10.1002/adhm.202000753] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/15/2020] [Indexed: 12/11/2022]
Abstract
Artificial nerve conduits capable of adequately releasing neurotrophic factors are extensively studied to bridge nerve defects. However, the lack of neurotrophic factors in the proximal area and their visible effects in axonal retrograde transport following nerve injury is one of the factors causing an incomplete nerve regeneration. Herein, an advanced conduit made of silk fibroin is produced, which can incorporate growth factors and promote an effective regeneration after injury. For that, enzymatically crosslinked silk fibroin-based conduits are developed to be used as a platform for the controlled delivery of neurotrophic factors. Nerve growth factor and glial-cell line derived neurotrophic factor (GDNF) are incorporated using two different methodologies: i) crosslinking and ii) absorption method. The release profile is measured by ELISA technique. The bioactivity of the neurotrophic factors is evaluated in vitro by using primary dorsal root ganglia. When implanted in a 10 mm sciatic nerve defect in rats, GDNF-loaded silk fibroin conduits reveal retrograde neuroprotection as compared to autografts and plain silk fibroin conduit. Therefore, the novel design presents a substantial improvement of retrograde trafficking, neurons' protection, and motor nerve reinnervation.
Collapse
Affiliation(s)
- Cristiana R. Carvalho
- 3B's Research Group‐Biomaterials Biodegradables and Biomimetics Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine University of Minho Barco Guimarães 4805‐017 Portugal
| | - Wei Chang
- New Jersey Center for Biomaterials Rutgers The State University of New Jersey Piscataway NJ 08854 USA
| | - Joana Silva‐Correia
- 3B's Research Group‐Biomaterials Biodegradables and Biomimetics Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine University of Minho Barco Guimarães 4805‐017 Portugal
| | - Rui L. Reis
- 3B's Research Group‐Biomaterials Biodegradables and Biomimetics Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine University of Minho Barco Guimarães 4805‐017 Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group‐Biomaterials Biodegradables and Biomimetics Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine University of Minho Barco Guimarães 4805‐017 Portugal
| | - Joachim Kohn
- New Jersey Center for Biomaterials Rutgers The State University of New Jersey Piscataway NJ 08854 USA
| |
Collapse
|
15
|
Eskandari N, Boroujeni ME, Abdollahifar MA, Piryaei A, Khodagholi F, Mirbehbahani SH, Siroosi S, Moghaddam MH, Aliaghaei A, Sadeghi Y. Transplantation of human dental pulp stem cells compensates for striatal atrophy and modulates neuro-inflammation in 3-nitropropionic acid rat model of Huntington's disease. Neurosci Res 2020; 170:133-144. [PMID: 33359180 DOI: 10.1016/j.neures.2020.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/09/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023]
Abstract
Stem cell-based therapy has recently offered a promising alternative for the remedy of neurodegenerative disorders like Huntington's disease (HD). Herein, we investigated the potential ameliorative effects of implantation of dental pulp stem cells (DPSCs) in 3-nitropropionic acid (3-NP) rat models of HD. In this regard, human DPSCs were isolated, culture-expanded and implanted in rats lesioned with 3-NP. Post-transplantation examinations revealed that DPSCs were able to survive and augment motor skills and muscle activity. Histological analysis showed DPSCs treatment hampered the shrinkage of the striatum along with the inhibition of gliosis and microgliosis in the striatum of 3-NP rat models. We also detected the downregulation of Caspase-3 and pro-inflammatory cytokines such as TNF and IL-1β upon DPSCs grafting. Overall, these findings imply that the grafting of DPSCs could repair motor-skill impairment and induce neurogenesis, probably through the secretion of neurotrophic factors and the modulation of neuroinflammatory response in HD animal models.
Collapse
Affiliation(s)
- Neda Eskandari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Mohammad Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shokoofeh Siroosi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Yousef Sadeghi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Anatomy & Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
16
|
Neurotrophic Factors in Glaucoma and Innovative Delivery Systems. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10249015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glaucoma is a neurodegenerative disease and a worldwide leading cause of irreversible vision loss. In the last decades, high efforts have been made to develop novel treatments effective in inducing protection and/or recovery of neural function in glaucoma, including neurotrophic factors (NTFs). These approaches have shown encouraging data in preclinical setting; however, the challenge of sustained, targeted delivery to the retina and optic nerve still prevents the clinical translation. In this paper, the authors review and discuss the most recent advances for the use of NTFs treatment in glaucoma, including intraocular delivery. Novel strategies in drug and gene delivery technology for NTFs are proving effective in promoting long-term retinal ganglion cells (RGCs) survival and related functional improvements. Results of experimental and clinical studies evaluating the efficacy and safety of biodegradable slow-release NTF-loaded microparticle devices, encapsulated NTF-secreting cells implants, mimetic ligands for NTF receptors, and viral and non-viral NTF gene vehicles are discussed. NTFs are able to prevent and even reverse apoptotic ganglion cell death. Nevertheless, neuroprotection in glaucoma remains an open issue due to the unmet need of sustained delivery to the posterior segment of the eye. The recent advances in intraocular delivery systems pave the way for possible future use of NTFs in clinical practice for the treatment of glaucoma.
Collapse
|
17
|
Sharif M, Noroozian M, Hashemian F. Do serum GDNF levels correlate with severity of Alzheimer's disease? Neurol Sci 2020; 42:2865-2872. [PMID: 33215334 DOI: 10.1007/s10072-020-04909-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION A growing body of evidence that glial cell line-derived neurotrophic factor (GDNF) levels are probably involved in pathogenesis and disease course of Alzheimer's disease (AD) suggested that its blood levels could potentially be used as a biomarker of AD. The aim of this study was to compare serum GDNF levels in patients with AD and age-matched controls. METHODS Serum concentrations of GDNF were compared in 25 AD patients and 25 healthy volunteers using a double-antibody sandwich enzyme-linked immunosorbent assay (ELISA). Severity of the disease in AD patients was assessed using Functional Assessment Staging (FAST). Cognitive assessment of the patients was done using the Mini-Mental State Examination (MMSE). RESULTS Mean GDNF levels were found to be 2.45 ± 0.93 ng/ml in AD patients and 4.61 ± 3.39 ng/ml in age-matched controls. There was a statistically significant difference in GDNF serum levels in patients with AD compared to age-matched controls (p = 0.001). Moreover, GDNF serum levels were significantly correlated with disease severity (p < 0.001) and cognitive impairment (p < 0.001). CONCLUSION This study showed that serum levels of GDNF are significantly decreased in AD patients in comparison with age-matched controls, thus suggesting a potential role of GDNF as a disease biomarker. However, a comprehensive study of changes in serum levels of multiple neurotrophic factors reflective of different neurobiological pathways in large-scale population studies is recommended.
Collapse
Affiliation(s)
- Maryam Sharif
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, 99 Yakhchal Street, Shariati Avenue, Tehran, 1941933111, Iran
| | - Maryam Noroozian
- Memory and Behavioral Neurology Division, Department of Psychiatry, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Hashemian
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, 99 Yakhchal Street, Shariati Avenue, Tehran, 1941933111, Iran.
| |
Collapse
|
18
|
Lovett ML, Nieland TJ, Dingle YTL, Kaplan DL. Innovations in 3-Dimensional Tissue Models of Human Brain Physiology and Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909146. [PMID: 34211358 PMCID: PMC8240470 DOI: 10.1002/adfm.201909146] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 05/04/2023]
Abstract
3-dimensional (3D) laboratory tissue cultures have emerged as an alternative to traditional 2-dimensional (2D) culture systems that do not recapitulate native cell behavior. The discrepancy between in vivo and in vitro tissue-cell-molecular responses impedes understanding of human physiology in general and creates roadblocks for the discovery of therapeutic solutions. Two parallel approaches have emerged for the design of 3D culture systems. The first is biomedical engineering methodology, including bioengineered materials, bioprinting, microfluidics and bioreactors, used alone or in combination, to mimic the microenvironments of native tissues. The second approach is organoid technology, in which stem cells are exposed to chemical and/or biological cues to activate differentiation programs that are reminiscent of human (prenatal) development. This review article describes recent technological advances in engineering 3D cultures that more closely resemble the human brain. The contributions of in vitro 3D tissue culture systems to new insights in neurophysiology, neurological diseases and regenerative medicine are highlighted. Perspectives on designing improved tissue models of the human brain are offered, focusing on an integrative approach merging biomedical engineering tools with organoid biology.
Collapse
Affiliation(s)
- Michael L. Lovett
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Thomas J.F. Nieland
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Yu-Ting L. Dingle
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| |
Collapse
|
19
|
Localized delivery of CRISPR/dCas9 via layer-by-layer self-assembling peptide coating on nanofibers for neural tissue engineering. Biomaterials 2020; 256:120225. [DOI: 10.1016/j.biomaterials.2020.120225] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
|
20
|
Yazdanian M, Moazzami M, Shabani M, Cheragh Birjandi S. The Effect of Eight-Week Aerobic Exercise before Cerebral Ischemia on the Expression of NT-3 and TrkC Genes in Male Rats. MEDICAL LABORATORY JOURNAL 2020. [DOI: 10.29252/mlj.14.5.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
21
|
Chen X, Xiaokaiti M, Wu S, Yao W, You S, Li G, Mo X. A modified fabrication procedure of retinal explant and optimized formulation of culture medium in a three-dimensional retinal culture system. J Neurosci Methods 2020; 344:108860. [PMID: 32673635 DOI: 10.1016/j.jneumeth.2020.108860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/23/2020] [Accepted: 07/10/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Three-dimensional culture system of retinal explant is commonly used to study retinal ganglion cell (RGC) axon regeneration in vitro. The retinal explants fabricated by traditional procedure in culture system, however, are usually too small (merely 0.5 × 0.5 mm) to be easily detected or treated by current experimental techniques. Also, the constituents of culture medium have not been fully elucidated. NEW METHOD A fabrication procedure was developed to enlarge the retinal explants and explore the reasonable concentration of fetal bovine serum (FBS) for evaluating axonal regeneration. RESULTS There were no significant differences in the density or length of regenerative neurites in the retinal explants fabricated by traditional and modified procedures. Increased FBS concentrations promoted neurite regeneration, decreased RGCs apoptosis, and activated tyrosine kinase B (TrkB) receptors, all reaching a plateau at 1 % FBS. COMPARISON WITH EXISTING METHODS Compared with traditional procedure, the modified fabrication procedure facilitates application of experimental techniques to retinal explants, increases the efficiency of obtaining observation area of regenerating neurites, and reduces the wastage of retinal tissues. The recommended FBS concentration determined in this study is shown to be more suitable for studying neuronal regeneration. CONCLUSION The retinal explants made by the modified fabrication procedure are successfully applied to the three-dimensional culture system, and presented several advantages over the traditional one. Furthermore, a preliminary experiment must be performed to determine the suitable concentration of FBS in each study to ensure accuracy and stability of the results obtained from the three-dimension retinal culture system.
Collapse
Affiliation(s)
- Xiangwu Chen
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China
| | - Maierhaba Xiaokaiti
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China
| | - Suqian Wu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China
| | - Wang Yao
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China
| | - Shuqi You
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China
| | - Gang Li
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China
| | - Xiaofen Mo
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, NO. 83, Fenyang Road, Shanghai, 200032, China.
| |
Collapse
|
22
|
Sandoval‐Castellanos AM, Claeyssens F, Haycock JW. Biomimetic surface delivery of NGF and BDNF to enhance neurite outgrowth. Biotechnol Bioeng 2020; 117:3124-3135. [DOI: 10.1002/bit.27466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 11/11/2022]
Affiliation(s)
| | - Frederik Claeyssens
- Department of Materials Science and Engineering The University of Sheffield Sheffield United Kingdom
| | - John W. Haycock
- Department of Materials Science and Engineering The University of Sheffield Sheffield United Kingdom
| |
Collapse
|
23
|
Fadia NB, Bliley JM, DiBernardo GA, Crammond DJ, Schilling BK, Sivak WN, Spiess AM, Washington KM, Waldner M, Liao HT, James IB, Minteer DM, Tompkins-Rhoades C, Cottrill AR, Kim DY, Schweizer R, Bourne DA, Panagis GE, Asher Schusterman M, Egro FM, Campwala IK, Simpson T, Weber DJ, Gause T, Brooker JE, Josyula T, Guevara AA, Repko AJ, Mahoney CM, Marra KG. Long-gap peripheral nerve repair through sustained release of a neurotrophic factor in nonhuman primates. Sci Transl Med 2020; 12:12/527/eaav7753. [DOI: 10.1126/scitranslmed.aav7753] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/26/2019] [Accepted: 11/25/2019] [Indexed: 01/09/2023]
Abstract
Severe injuries to peripheral nerves are challenging to repair. Standard-of-care treatment for nerve gaps >2 to 3 centimeters is autografting; however, autografting can result in neuroma formation, loss of sensory function at the donor site, and increased operative time. To address the need for a synthetic nerve conduit to treat large nerve gaps, we investigated a biodegradable poly(caprolactone) (PCL) conduit with embedded double-walled polymeric microspheres encapsulating glial cell line–derived neurotrophic factor (GDNF) capable of providing a sustained release of GDNF for >50 days in a 5-centimeter nerve defect in a rhesus macaque model. The GDNF-eluting conduit (PCL/GDNF) was compared to a median nerve autograft and a PCL conduit containing empty microspheres (PCL/Empty). Functional testing demonstrated similar functional recovery between the PCL/GDNF-treated group (75.64 ± 10.28%) and the autograft-treated group (77.49 ± 19.28%); both groups were statistically improved compared to PCL/Empty-treated group (44.95 ± 26.94%). Nerve conduction velocity 1 year after surgery was increased in the PCL/GDNF-treated macaques (31.41 ± 15.34 meters/second) compared to autograft (25.45 ± 3.96 meters/second) and PCL/Empty (12.60 ± 3.89 meters/second) treatment. Histological analyses included assessment of Schwann cell presence, myelination of axons, nerve fiber density, and g-ratio. PCL/GDNF group exhibited a statistically greater average area occupied by individual Schwann cells at the distal nerve (11.60 ± 33.01 μm2) compared to autograft (4.62 ± 3.99 μm2) and PCL/Empty (4.52 ± 5.16 μm2) treatment groups. This study demonstrates the efficacious bridging of a long peripheral nerve gap in a nonhuman primate model using an acellular, biodegradable nerve conduit.
Collapse
Affiliation(s)
- Neil B. Fadia
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jacqueline M. Bliley
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Donald J. Crammond
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Wesley N. Sivak
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alexander M. Spiess
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kia M. Washington
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Matthias Waldner
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Han-Tsung Liao
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Isaac B. James
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Danielle M. Minteer
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Adam R. Cottrill
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Deok-Yeol Kim
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Riccardo Schweizer
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Debra A. Bourne
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - George E. Panagis
- Department of Biology, University of Pittsburgh, Greensburg, PA 15601, USA
| | - M. Asher Schusterman
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Francesco M. Egro
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Tyler Simpson
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Douglas J. Weber
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Trent Gause
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jack E. Brooker
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tvisha Josyula
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Astrid A. Guevara
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alexander J. Repko
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Kacey G. Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
24
|
Carvalho CR, Oliveira JM, Reis RL. Modern Trends for Peripheral Nerve Repair and Regeneration: Beyond the Hollow Nerve Guidance Conduit. Front Bioeng Biotechnol 2019; 7:337. [PMID: 31824934 PMCID: PMC6882937 DOI: 10.3389/fbioe.2019.00337] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve repair and regeneration remains among the greatest challenges in tissue engineering and regenerative medicine. Even though peripheral nerve injuries (PNIs) are capable of some degree of regeneration, frail recovery is seen even when the best microsurgical technique is applied. PNIs are known to be very incapacitating for the patient, due to the deprivation of motor and sensory abilities. Since there is no optimal solution for tackling this problem up to this day, the evolution in the field is constant, with innovative designs of advanced nerve guidance conduits (NGCs) being reported every day. As a basic concept, a NGC should act as a physical barrier from the external environment, concomitantly acting as physical guidance for the regenerative axons across the gap lesion. NGCs should also be able to retain the naturally released nerve growth factors secreted by the damaged nerve stumps, as well as reducing the invasion of scar tissue-forming fibroblasts to the injury site. Based on the neurobiological knowledge related to the events that succeed after a nerve injury, neuronal subsistence is subjected to the existence of an ideal environment of growth factors, hormones, cytokines, and extracellular matrix (ECM) factors. Therefore, it is known that multifunctional NGCs fabricated through combinatorial approaches are needed to improve the functional and clinical outcomes after PNIs. The present work overviews the current reports dealing with the several features that can be used to improve peripheral nerve regeneration (PNR), ranging from the simple use of hollow NGCs to tissue engineered intraluminal fillers, or to even more advanced strategies, comprising the molecular and gene therapies as well as cell-based therapies.
Collapse
Affiliation(s)
- Cristiana R. Carvalho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| |
Collapse
|
25
|
Yazdanian M, Moazami M, Shabani M, Cheragh Birjandi S. Effects of Exercise Preconditioning on Neurotrophin-4 and Tropomyosin Receptor Kinase B Expression in the Hippocampal CA1 Region Following Transient Global Cerebral Ischemia/Reperfusion in Wistar Rats. MEDICAL LABORATORY JOURNAL 2019. [DOI: 10.29252/mlj.13.6.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
26
|
Moradi P, Ganjkhani M, Anarkooli IJ, Abdanipour A. Neuroprotective effects of lovastatin in the pilocarpine rat model of epilepsy according to the expression of neurotrophic factors. Metab Brain Dis 2019; 34:1061-1069. [PMID: 31144103 DOI: 10.1007/s11011-019-00424-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/22/2019] [Indexed: 12/18/2022]
Abstract
Studies have suggested that neurotrophic factors (NTFs) are involved in the status epilepticus development. This indicates their essential role in mediating acquired epileptic conditions. Therefore, modulating the expression of NTFs may inhibit seizure-induced cell loss in the epileptic lesions. In this study, we examined the anti-apoptotic, anti-necrotic and regulatory effects of lovastatin on the expression of NTFs in the pilocarpine rat model of temporal lobe epilepsy (TLE). A total of 32 male Wistar rats were divided into 4 groups (n = 8 per group): i) normal; ii) non-treated epileptic group [intraperitoneal (i.p.) administration of 350-400 mg/kg pilocarpine]; iii) treatment group (pilocarpine-treated rats treated followed by 5 mg/kg lovastatin); and iv) vehicle epileptic rats treated with Carboxymethyl cellulose (CMC). Animals that had a behavioral score of 4-5 according to the Racine scale were selected for study participation. Three days after the first seizure, pilocarpine-treated rats received i.p. injections of lovastatin for 14 days. The rats were killed and prepared for histopathologic analysis as well as real-time RT-PCR 17 days after the first seizure. The results of this study showed increased mRNA expression of glial cell line-derived neurotrophic factor (GDNF) and Ciliary neurotrophic factor (CNTF) and decreased expressions of Brain-derived neurotrophic factor (BDNF), Neurotrophin-3 (NT-3), and Neurotrophin-4 (NT-4) mRNA in the epileptic rats treated with lovastatin. Histological analysis of neurodegeneration in the brain sections showed that the number of hippocampal apoptotic and necrotic cells significantly decreased in the treatment groups. Furthermore, numerical density of neurons per area was significantly higher in the treated groups compared with the untreated groups. Collectively, the results of this study have shown that lovastatin could attenuate seizure-induced expression of neurotrophic factors and consequently reduce hippocampal cell death in the pilocarpine rat model of TLE.
Collapse
Affiliation(s)
- Pooyan Moradi
- Department of Physiology and Pharmacology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), P.O. Box 45139-56184, Zanjan, Iran
| | - Mahin Ganjkhani
- Department of Physiology and Pharmacology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), P.O. Box 45139-56184, Zanjan, Iran.
| | - Iraj Jafari Anarkooli
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences (ZUMS), P.O. Box 45139-56184, Zanjan, Iran
| | - Alireza Abdanipour
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences (ZUMS), P.O. Box 45139-56184, Zanjan, Iran.
| |
Collapse
|
27
|
Fogli B, Corthout N, Kerstens A, Bosse F, Klimaschewski L, Munck S, Schweigreiter R. Imaging axon regeneration within synthetic nerve conduits. Sci Rep 2019; 9:10095. [PMID: 31300753 PMCID: PMC6626049 DOI: 10.1038/s41598-019-46579-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/28/2019] [Indexed: 11/23/2022] Open
Abstract
While axons within the central nervous system (CNS) do not regenerate following injury, those in the peripheral nervous system (PNS) do, although not in a clinically satisfactory manner as only a small proportion of axons exhibit long-distance regeneration. Moreover, functional recovery is hampered by excessive axonal sprouting and aberrant reinnervation of target tissue. In order to investigate the mechanisms governing the regrowth of axons following injury, previous studies have used lesion paradigms of peripheral nerves in rat or mouse models, and reagents or cells have been administered to the lesion site through nerve conduits, aiming to improve early-stage regeneration. Morphological analysis of such in vivo experiments has however been limited by the incompatibility of synthetic nerve conduits with existing tissue-clearing and imaging techniques. We present herein a novel experimental approach that allows high-resolution imaging of individual axons within nerve conduits, together with quantitative assessment of fiber growth. We used a GFP-expressing mouse strain in a lesion model of the sciatic nerve to describe a strategy that combines nerve clearing, chemical treatment of chitosan nerve conduits, and long working distance confocal microscopy with image processing and analysis. This novel experimental setup provides a means of documenting axon growth within the actual conduit during the critical initial stage of regeneration. This will greatly facilitate the development and evaluation of treatment regimens to improve axonal regeneration following nerve damage.
Collapse
Affiliation(s)
- Barbara Fogli
- Innsbruck Medical University, Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, 6020, Innsbruck, Austria
| | - Nikky Corthout
- VIB-KU Leuven Center for Brain & Disease Research O&N 4, Campus Gasthuisberg, 3000, Leuven, Belgium.,KU Leuven, Department for Neuroscience, Campus Gasthuisberg, 3000, Leuven, Belgium.,VIB Bio Imaging Core, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Axelle Kerstens
- VIB-KU Leuven Center for Brain & Disease Research O&N 4, Campus Gasthuisberg, 3000, Leuven, Belgium.,KU Leuven, Department for Neuroscience, Campus Gasthuisberg, 3000, Leuven, Belgium.,VIB Bio Imaging Core, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Frank Bosse
- Heinrich-Heine-University Düsseldorf, Department of Neurology, Molecular Neurobiology Laboratory, 40225, Düsseldorf, Germany
| | - Lars Klimaschewski
- Innsbruck Medical University, Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, 6020, Innsbruck, Austria
| | - Sebastian Munck
- VIB-KU Leuven Center for Brain & Disease Research O&N 4, Campus Gasthuisberg, 3000, Leuven, Belgium. .,KU Leuven, Department for Neuroscience, Campus Gasthuisberg, 3000, Leuven, Belgium. .,VIB Bio Imaging Core, Campus Gasthuisberg, 3000, Leuven, Belgium.
| | - Rüdiger Schweigreiter
- Innsbruck Medical University, Biocenter, Division of Neurobiochemistry, 6020, Innsbruck, Austria.
| |
Collapse
|
28
|
Eltahir HM. Insight into the roles of hepatoma derived growth factor related protein-3 under physiological and pathological conditions. Biochem Cell Biol 2018; 96:707-712. [DOI: 10.1139/bcb-2018-0098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hepatoma derived growth factor related protein-3 (HRP-3) is a HDGF growth factor family member that is expressed mainly in nervous tissues. It shares structural similarities with HDGF but differs in its expression and scope of action. It has recently attracted more attention due to its variable roles. HRP-3 was originally reported as a mitogenic factor. However, over the last decade, additional functions for this growth factor have been uncovered. In addition to its mitogenic activity, other physiological functions were discovered including those related to proliferation, differentiation, and maintenance of neurons, presenting it as a neurotrophic and neuroprotective growth factor. Interestingly, HRP-3 was also shown to be involved in the pathogenesis of certain tumors via its mitogenic, angiogenic, and antiapoptotic effects. Based on this, HRP-3 represents a molecule that can be targeted for the treatment of cancer and various neurodegenerative diseases.
Collapse
Affiliation(s)
- Heba M. Eltahir
- College of Pharmacy, Department of Pharmacology and Toxicology, Taibah University, Medina, KSA
- College of Pharmacy, Department of Pharmacology and Toxicology, Taibah University, Medina, KSA
| |
Collapse
|
29
|
Alqawlaq S, Flanagan JG, Sivak JM. All roads lead to glaucoma: Induced retinal injury cascades contribute to a common neurodegenerative outcome. Exp Eye Res 2018; 183:88-97. [PMID: 30447198 DOI: 10.1016/j.exer.2018.11.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
Abstract
Glaucoma describes a distinct optic neuropathy with complex etiology and a variety of associated risk factors, but with similar pathological endpoints. Risk factors such as age, increased intraocular pressure (IOP), low mean arterial pressure, and autoimmune disease, can all be associated with death of retinal ganglion cells (RGCs) and optic nerve head remodeling. Today, IOP management remains the standard of care, even though IOP elevation is not pathognomonic of glaucoma, and patients can continue to lose vision despite effective IOP control. A contemporary view of glaucoma as a complex, neurodegenerative disease has developed, along with the recognition of a need for new disease modifying retinal treatment strategies and improved outcomes. However, the distinction between risk factors triggering the disease process and retinal injury responses is not always clear. In this review, we attempt to distinguish between the various triggers, and their association with subsequent key RGC injury mechanisms. We propose that distinct glaucomatous risk factors result in similar retinal and optic nerve injury cascades, including oxidative and metabolic stress, glial reactivity, and altered inflammatory responses, which induce common molecular signals to induce RGC apoptosis. This organization forms a coherent disease framework and presents conserved targets for therapeutic intervention that are not limited to specific risk factors.
Collapse
Affiliation(s)
- Samih Alqawlaq
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - John G Flanagan
- School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Neurotrophic Activity of the Carrageenophyte Kappaphycus alvarezii Cultivated at Different Depths and for Different Growth Periods in Various Areas of Indonesia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1098076. [PMID: 30420890 PMCID: PMC6211153 DOI: 10.1155/2018/1098076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/17/2018] [Accepted: 09/24/2018] [Indexed: 11/22/2022]
Abstract
The carrageenophyte Kappaphycus alvarezii (Rhodophyta) has neurotrophic activity in primary hippocampal neurons. This seaweed is abundant and easily cultivated in tropical coastal areas. To determine the best growth conditions for neurotrophic activity, thalli were grown at different depths and for different periods in various areas of Indonesia. Neurotrophic activity was measured based on the number of primary neurites, the total length of the primary neurites, and the length of the longest neurite. K. alvarezii had higher neurotrophic activity than carrageenophytes K. striatum and Eucheuma denticulatum cultured under the same conditions. K. alvarezii grown at the surface for 45 days had higher (1.4- to 1.8-fold) neurotrophic activity than thalli grown at depth (2 m) or harvested sooner (15 days) (P < 0.05). Relatively high activities were detected in thalli cultured at Ternate and Garut, Indonesia. Therefore, from a commercial perspective, the culture conditions at the surface for 45 days were optimal for the production of both neurotrophic compounds and carrageenan. K. alvarezii produced neurotrophic compounds under various environmental conditions, although some conditions were optimal.
Collapse
|
31
|
Valvassori SS, Borges C, Bavaresco DV, Varela RB, Resende WR, Peterle BR, Arent CO, Budni J, Quevedo J. Hypericum perforatum chronic treatment affects cognitive parameters and brain neurotrophic factor levels. ACTA ACUST UNITED AC 2018; 40:367-375. [PMID: 30110089 PMCID: PMC6899380 DOI: 10.1590/1516-4446-2017-2271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/21/2017] [Indexed: 12/21/2022]
Abstract
Objective: To evaluate the effects of Hypericum perforatum (hypericum) on cognitive behavior and neurotrophic factor levels in the brain of male and female rats. Methods: Male and female Wistar rats were treated with hypericum or water during 28 days by gavage. The animals were then subjected to the open-field test, novel object recognition and step-down inhibitory avoidance test. Nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and glial cell-line derived neurotrophic factor (GDNF) levels were evaluated in the hippocampus and frontal cortex. Results: Hypericum impaired the acquisition of short- and long-term aversive memory in male rats, evaluated in the inhibitory avoidance test. Female rats had no immediate memory acquisition and decreased short-term memory acquisition in the inhibitory avoidance test. Hypericum also decreased the recognition index of male rats in the object recognition test. Female rats did not recognize the new object in either the short-term or the long-term memory tasks. Hypericum decreased BDNF in the hippocampus of male and female rats. Hypericum also decreased NGF in the hippocampus of female rats. Conclusions: The long-term administration of hypericum appears to cause significant cognitive impairment in rats, possibly through a reduction in the levels of neurotrophic factors. This effect was more expressive in females than in males.
Collapse
Affiliation(s)
- Samira S Valvassori
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.,Laboratório de Sinalização Neural e Psicofarmacologia, PPGCS, UNASAU, UNESC, Criciúma, SC, Brazil
| | - Cenita Borges
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - Daniela V Bavaresco
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.,Laboratório de Sinalização Neural e Psicofarmacologia, PPGCS, UNASAU, UNESC, Criciúma, SC, Brazil
| | - Roger B Varela
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.,Laboratório de Sinalização Neural e Psicofarmacologia, PPGCS, UNASAU, UNESC, Criciúma, SC, Brazil
| | - Wilson R Resende
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.,Laboratório de Sinalização Neural e Psicofarmacologia, PPGCS, UNASAU, UNESC, Criciúma, SC, Brazil
| | - Bruna R Peterle
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.,Laboratório de Sinalização Neural e Psicofarmacologia, PPGCS, UNASAU, UNESC, Criciúma, SC, Brazil
| | - Camila O Arent
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - Josiane Budni
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Unidade Acadêmica de Ciências da Saúde (UNASAU), Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.,Department of Psychiatry and Behavioral Sciences, Center for Experimental Models in Psychiatry, The University of Texas Medical School at Houston, Houston, TX, USA
| |
Collapse
|
32
|
Mòdol-Caballero G, Santos D, Navarro X, Herrando-Grabulosa M. Neuregulin 1 Reduces Motoneuron Cell Death and Promotes Neurite Growth in an in Vitro Model of Motoneuron Degeneration. Front Cell Neurosci 2018; 11:431. [PMID: 29375317 PMCID: PMC5767462 DOI: 10.3389/fncel.2017.00431] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disorder with no effective treatment currently available. Although the mechanisms of motoneuron (MN) death are still unclear, glutamate excitotoxicity and neuroinflammatory reaction are two main features in the neurodegenerative process of ALS. Neuregulin 1 (NRG1) is a trophic factor highly expressed in MNs and neuromuscular junctions. Several recent evidences suggest that NRG1 and their ErbB receptors are involved in ALS. However, further knowledge is still needed to clarify the role of the NRG1-ErbB pathway on MN survival. In this study we used an in vitro model of spinal cord organotypic cultures (SCOCs) subject to chronic excitotoxicity caused by DL-threo-β-hydroxyaspartic acid (THA) to characterize the effect of NRG1 on MN survival. Our results show that addition of recombinant human NRG1 (rhNRG1) to the medium significantly increased MN survival through the activation of ErbB receptors which was ablated with lapatinib (LP), an ErbB inhibitor, and reduced microglial reactivity overcoming the excitotoxicity effects. rhNRG1 activated the pro-survival PI3K/AKT pathway and restored the autophagic flux in the spinal cord culture. Moreover, addition of rhNRG1 to the medium promoted motor and sensory neurite outgrowth. These findings indicate that increasing NRG1 at the spinal cord is an interesting approach for promoting MN protection and regeneration.
Collapse
Affiliation(s)
- Guillem Mòdol-Caballero
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Daniel Santos
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Mireia Herrando-Grabulosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| |
Collapse
|
33
|
Hsu MN, Liao HT, Li KC, Chen HH, Yen TC, Makarevich P, Parfyonova Y, Hu YC. Adipose-derived stem cell sheets functionalized by hybrid baculovirus for prolonged GDNF expression and improved nerve regeneration. Biomaterials 2017; 140:189-200. [DOI: 10.1016/j.biomaterials.2017.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 01/12/2023]
|
34
|
McIsaac W, Ferguson AV. Glucose concentrations modulate brain-derived neurotrophic factor responsiveness of neurones in the paraventricular nucleus of the hypothalamus. J Neuroendocrinol 2017; 29. [PMID: 28258626 DOI: 10.1111/jne.12464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/07/2017] [Accepted: 02/20/2017] [Indexed: 11/30/2022]
Abstract
The hypothalamic paraventricular nucleus (PVN) is critical for normal energy balance and has been shown to contain high levels of both brain-derived neurotrophic factor (BDNF) and tropomyosin-receptor kinase B mRNA. Microinjections of BDNF into the PVN increase energy expenditure, suggesting that BDNF plays an important role in energy homeostasis through direct actions in this nucleus. The present study aimed to examine the postsynaptic effects of BDNF on the membrane potential of PVN neurones, and also to determine whether extracellular glucose concentrations modulated these effects. We used hypothalamic PVN slices from male Sprague-Dawley rats to perform whole cell current-clamp recordings from PVN neurones. BDNF was bath applied at a concentration of 2 nmol L-1 and the effects on membrane potential determined. BDNF caused depolarisations in 54% of neurones (n=25; mean±SEM, 8.9±1.2 mV) and hyperpolarisations in 23% (n=11; -6.7±1.4 mV), whereas the remaining cells were unaffected. These effects were maintained in the presence of tetrodotoxin (n=9; 56% depolarised, 22% hyperpolarised, 22% nonresponders), or the GABAa antagonist bicuculline (n=12; 42% depolarised, 17% hyperpolarised, 41% nonresponders), supporting the conclusion that these effects on membrane potential were postsynaptic. Current-clamp recordings from PVN neurones next examined the effects of BDNF on these neurones at varying extracellular glucose concentrations. Larger proportions of PVN neurones hyperpolarised in response to BDNF as the glucose concentrations decreased [10 mmol L-1 glucose 23% (n=11) of neurones hyperpolarised, whereas, at 0.2 mmol L-1 glucose, 71% showed hyperpolarising effects (n=12)]. Our findings reveal that BDNF has direct GABAA independent effects on PVN neurones, which are modulated by local glucose concentrations. The latter observation further emphasises the critical importance of using physiologically relevant conditions in an investigation of the central pathways involved in the regulation of energy homeostasis.
Collapse
Affiliation(s)
- W McIsaac
- Centre for Neuroscience, Queens University, Kingston, ON, Canada
| | - A V Ferguson
- Centre for Neuroscience, Queens University, Kingston, ON, Canada
| |
Collapse
|
35
|
Tang T, Li Y, Jiao Q, Du X, Jiang H. Cerebral Dopamine Neurotrophic Factor: A Potential Therapeutic Agent for Parkinson's Disease. Neurosci Bull 2017; 33:568-575. [PMID: 28337696 DOI: 10.1007/s12264-017-0123-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/18/2016] [Indexed: 11/27/2022] Open
Abstract
The application of neurotrophic factors (NTFs) is a promising therapeutic strategy for neurodegenerative disorders such as Parkinson's disease (PD). Many NTFs have been reported to enhance the survival, regeneration, and differentiation of neurons and to induce synaptic plasticity. However, because of their potential side-effects and low efficacy after clinical administration, more potent treatments for neurodegenerative disorders are being sought. Cerebral dopamine neurotrophic factor (CDNF), a newly-identified NTF homologous to mesencephalic astrocyte-derived NTF, is structurally and functionally different from other NTFs, providing new hope especially for PD patients. In various animal models of PD, CDNF is efficient in protecting and repairing dopaminergic neurons, and it inhibits endoplasmic reticulum stress, neuroinflammation, and apoptosis. Recent progress in all facets of CDNF research has enabled researchers to better understand its beneficial effects in the treatment of PD.
Collapse
Affiliation(s)
- Tingting Tang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
36
|
Xu F, Zhang K, Lv P, Lu R, Zheng L, Zhao J. NECL1 coated PLGA as favorable conduits for repair of injured peripheral nerve. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:1132-1140. [DOI: 10.1016/j.msec.2016.03.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/26/2016] [Accepted: 03/14/2016] [Indexed: 01/13/2023]
|
37
|
Fadda A, Bärtschi M, Hemphill A, Widmer HR, Zurbriggen A, Perona P, Vidondo B, Oevermann A. Primary Postnatal Dorsal Root Ganglion Culture from Conventionally Slaughtered Calves. PLoS One 2016; 11:e0168228. [PMID: 27936156 PMCID: PMC5148591 DOI: 10.1371/journal.pone.0168228] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
Neurological disorders in ruminants have an important impact on veterinary health, but very few host-specific in vitro models have been established to study diseases affecting the nervous system. Here we describe a primary neuronal dorsal root ganglia (DRG) culture derived from calves after being conventionally slaughtered for food consumption. The study focuses on the in vitro characterization of bovine DRG cell populations by immunofluorescence analysis. The effects of various growth factors on neuron viability, neurite outgrowth and arborisation were evaluated by morphological analysis. Bovine DRG neurons are able to survive for more than 4 weeks in culture. GF supplementation is not required for neuronal survival and neurite outgrowth. However, exogenously added growth factors promote neurite outgrowth. DRG cultures from regularly slaughtered calves represent a promising and sustainable host specific model for the investigation of pain and neurological diseases in bovines.
Collapse
Affiliation(s)
- A. Fadda
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, Theodor Kocher Institute, University of Bern, Switzerland
| | - M. Bärtschi
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - A. Hemphill
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - H. R. Widmer
- Neurocenter and Regenerative Neuroscience Cluster, University Hospital and University of Bern, Bern, Switzerland
| | - A. Zurbriggen
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - P. Perona
- School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| | - B. Vidondo
- Veterinary Public Health Institute (VPHI), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - A. Oevermann
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
38
|
Sidorova YA, Saarma M. Glial cell line-derived neurotrophic factor family ligands and their therapeutic potential. Mol Biol 2016. [DOI: 10.1134/s0026893316040105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
39
|
Ciliary Neurotrophic Factor Promotes the Migration of Corneal Epithelial Stem/progenitor Cells by Up-regulation of MMPs through the Phosphorylation of Akt. Sci Rep 2016; 6:25870. [PMID: 27174608 PMCID: PMC4865747 DOI: 10.1038/srep25870] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/25/2016] [Indexed: 01/20/2023] Open
Abstract
The migration of limbal epithelial stem cells is important for the homeostasis and regeneration of corneal epithelium. Ciliary neurotrophic factor (CNTF) has been found to promote corneal epithelial wound healing by activating corneal epithelial stem/progenitor cells. However, the possible effect of CNTF on the migration of corneal epithelial stem/progenitor cells is not clear. This study found the expression of CNTF in mouse corneal epithelial stem/progenitor cells (TKE2) to be up-regulated after injury, on both gene and protein level. CNTF promoted migration of TKE2 in a dose-dependent manner and the peak was seen at 10 ng/ml. The phosphorylation level of Akt (p-Akt), and the expression of MMP3 and MMP14, were up-regulated after CNTF treatment both in vitro and in vivo. Akt and MMP3 inhibitor treatment delayed the migration effect by CNTF. Finally, a decreased expression of MMP3 and MMP14 was observed when Akt inhibitor was applied both in vitro and in vivo. This study provides new insights into the role of CNTF on the migration of corneal epithelial stem/progenitor cells and its inherent mechanism of Up-regulation of matrix metalloproteinases through the Akt signalling pathway.
Collapse
|
40
|
Wichmann H, Brinkhoff T, Simon M, Richter-Landsberg C. Dimethylsulfoniopropionate Promotes Process Outgrowth in Neural Cells and Exerts Protective Effects against Tropodithietic Acid. Mar Drugs 2016; 14:md14050089. [PMID: 27164116 PMCID: PMC4882563 DOI: 10.3390/md14050089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/22/2016] [Accepted: 04/27/2016] [Indexed: 01/10/2023] Open
Abstract
The marine environment harbors a plethora of bioactive substances, including drug candidates of potential value in the field of neuroscience. The present study was undertaken to investigate the effects of dimethylsulfoniopropionate (DMSP), produced by several algae, corals and higher plants, on cells of the mammalian nervous system, i.e., neuronal N2a and OLN-93 cells as model system for nerve cells and glia, respectively. Additionally, the protective capabilities of DMSP were assessed in cells treated with tropodithietic acid (TDA), a marine metabolite produced by several Roseobacter clade bacteria. Both cell lines, N2a and OLN-93, have previously been shown to be a sensitive target for the action of TDA, and cytotoxic effects of TDA have been connected to the induction of oxidative stress. Our data shows that DMSP promotes process outgrowth and microtubule reorganization and bundling, accompanied by an increase in alpha-tubulin acetylation. Furthermore, DMSP was able to prevent the cytotoxic effects exerted by TDA, including the breakdown of the mitochondrial membrane potential, upregulation of heat shock protein Hsp32 and activation of the extracellular signal-regulated kinases 1/2 (ERK1/2). Our study points to the conclusion that DMSP provides an antioxidant defense, not only in algae but also in mammalian neural cells.
Collapse
Affiliation(s)
- Heidi Wichmann
- Aquatic Microbial Ecology Group, Institute for Chemistry and Biology of the Marine Environment (ICBM), University of Oldenburg, Oldenburg 26129, Germany.
| | - Thorsten Brinkhoff
- Aquatic Microbial Ecology Group, Institute for Chemistry and Biology of the Marine Environment (ICBM), University of Oldenburg, Oldenburg 26129, Germany.
| | - Meinhard Simon
- Aquatic Microbial Ecology Group, Institute for Chemistry and Biology of the Marine Environment (ICBM), University of Oldenburg, Oldenburg 26129, Germany.
| | | |
Collapse
|
41
|
Wei L, Liu S, Conroy J, Wang J, Papanicolau-Sengos A, Glenn ST, Murakami M, Liu L, Hu Q, Conroy J, Miles KM, Nowak DE, Liu B, Qin M, Bshara W, Omilian AR, Head K, Bianchi M, Burgher B, Darlak C, Kane J, Merzianu M, Cheney R, Fabiano A, Salerno K, Talati C, Khushalani NI, Trump DL, Johnson CS, Morrison CD. Whole-genome sequencing of a malignant granular cell tumor with metabolic response to pazopanib. Cold Spring Harb Mol Case Stud 2016; 1:a000380. [PMID: 27148567 PMCID: PMC4850888 DOI: 10.1101/mcs.a000380] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Granular cell tumors are an uncommon soft tissue neoplasm. Malignant granular cell tumors comprise <2% of all granular cell tumors, are associated with aggressive behavior and poor clinical outcome, and are poorly understood in terms of tumor etiology and systematic treatment. Because of its rarity, the genetic basis of malignant granular cell tumor remains unknown. We performed whole-genome sequencing of one malignant granular cell tumor with metabolic response to pazopanib. This tumor exhibited a very low mutation rate and an overall stable genome with local complex rearrangements. The mutation signature was dominated by C>T transitions, particularly when immediately preceded by a 5' G. A loss-of-function mutation was detected in a newly recognized tumor suppressor candidate, BRD7. No mutations were found in known targets of pazopanib. However, we identified a receptor tyrosine kinase pathway mutation in GFRA2 that warrants further evaluation. To the best of our knowledge, this is only the second reported case of a malignant granular cell tumor exhibiting a response to pazopanib, and the first whole-genome sequencing of this uncommon tumor type. The findings provide insight into the genetic basis of malignant granular cell tumors and identify potential targets for further investigation.
Collapse
Affiliation(s)
- Lei Wei
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Jeffrey Conroy
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | - Sean T Glenn
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Mitsuko Murakami
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Lu Liu
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Jacob Conroy
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Kiersten Marie Miles
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - David E Nowak
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Biao Liu
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Maochun Qin
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Angela R Omilian
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Karen Head
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Michael Bianchi
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Blake Burgher
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Christopher Darlak
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - John Kane
- Department of Radiation Oncology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Mihai Merzianu
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Richard Cheney
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Andrew Fabiano
- Department of Surgery, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Kilian Salerno
- Department of Radiation Oncology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Chetasi Talati
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Nikhil I Khushalani
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Donald L Trump
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA;; Inova Dwight and Martha Schar Cancer Institute, Falls Church, Virginia 22042, USA
| | - Candace S Johnson
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | - Carl D Morrison
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| |
Collapse
|
42
|
The pruritus- and TH2-associated cytokine IL-31 promotes growth of sensory nerves. J Allergy Clin Immunol 2016; 138:500-508.e24. [PMID: 27212086 DOI: 10.1016/j.jaci.2016.02.020] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 01/25/2016] [Accepted: 02/04/2016] [Indexed: 01/27/2023]
Abstract
BACKGROUND Pruritus is a cardinal symptom of atopic dermatitis, and an increased cutaneous sensory network is thought to contribute to pruritus. Although the immune cell-IL-31-neuron axis has been implicated in severe pruritus during atopic skin inflammation, IL-31's neuropoietic potential remains elusive. OBJECTIVE We sought to analyze the IL-31-related transcriptome in sensory neurons and to investigate whether IL-31 promotes sensory nerve fiber outgrowth. METHODS In vitro primary sensory neuron culture systems were subjected to whole-transcriptome sequencing, ingenuity pathway analysis, immunofluorescence, and nerve elongation, as well as branching assays after IL-31 stimulation. In vivo we investigated the cutaneous sensory neuronal network in wild-type, Il31-transgenic, and IL-31 pump-equipped mice. RESULTS Transgenic Il31 overexpression and subcutaneously delivered IL-31 induced an increase in the cutaneous nerve fiber density in lesional skin in vivo. Transcriptional profiling of IL-31-activated dorsal root ganglia neurons revealed enrichment for genes promoting nervous system development and neuronal outgrowth and negatively regulating cell death. Moreover, the growth cones of primary small-diameter dorsal root ganglia neurons showed abundant IL-31 receptor α expression. Indeed, IL-31 selectively promoted nerve fiber extension only in small-diameter neurons. Signal transducer and activator of transcription 3 phosphorylation mediated IL-31-induced neuronal outgrowth, and pharmacologic inhibition of signal transducer and activator of transcription 3 completely abolished this effect. In contrast, transient receptor potential cation channel vanilloid subtype 1 channels were dispensable for IL-31-induced neuronal sprouting. CONCLUSIONS The pruritus- and TH2-associated novel cytokine IL-31 induces a distinct transcriptional program in sensory neurons, leading to nerve elongation and branching both in vitro and in vivo. This finding might help us understand the clinical observation that patients with atopic dermatitis experience increased sensitivity to minimal stimuli inducing sustained itch.
Collapse
|
43
|
Hsieh FY, Lin HH, Hsu SH. 3D bioprinting of neural stem cell-laden thermoresponsive biodegradable polyurethane hydrogel and potential in central nervous system repair. Biomaterials 2015; 71:48-57. [DOI: 10.1016/j.biomaterials.2015.08.028] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/13/2015] [Accepted: 08/14/2015] [Indexed: 01/14/2023]
|
44
|
Budni J, Bellettini-Santos T, Mina F, Garcez ML, Zugno AI. The involvement of BDNF, NGF and GDNF in aging and Alzheimer's disease. Aging Dis 2015; 6:331-41. [PMID: 26425388 DOI: 10.14336/ad.2015.0825] [Citation(s) in RCA: 263] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022] Open
Abstract
Aging is a normal physiological process accompanied by cognitive decline. This aging process has been the primary risk factor for development of aging-related diseases such as Alzheimer's disease (AD). Cognitive deficit is related to alterations of neurotrophic factors level such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and glial cell-derived neurotrophic factor (GDNF). These strong relationship between aging and AD is important to investigate the time which they overlap, as well as, the pathophysiological mechanism in each event. Considering that aging and AD are related to cognitive impairment, here we discuss the involving these neurotrophic factors in the aging process and AD.
Collapse
Affiliation(s)
- Josiane Budni
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Tatiani Bellettini-Santos
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Francielle Mina
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Michelle Lima Garcez
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Alexandra Ioppi Zugno
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
45
|
de Almeida JFA, Chen P, Henry MA, Diogenes A. Stem cells of the apical papilla regulate trigeminal neurite outgrowth and targeting through a BDNF-dependent mechanism. Tissue Eng Part A 2015; 20:3089-100. [PMID: 24837134 DOI: 10.1089/ten.tea.2013.0347] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Regenerative endodontic procedures have become a valuable alternative for the treatment of immature teeth with pulp necrosis. In addition to resolution of periradicular pathosis and promotion of continued root development, positive vitality testing has been observed in some regenerative clinical cases. Importantly, the positive response to electric stimulation of the regenerated tissue requires targeting of periradicular axons into the previously empty root canal space. However, the mechanism by which this process occurs is largely unknown. Since stem cells of the apical papilla (SCAP) have been proposed to populate the root canal following regenerative endodontic procedures, we hypothesized that SCAP regulate neurite outgrowth and axonal targeting. To test this hypothesis, we established primary co-cultures of human SCAP and rat trigeminal neurons, and performed neurite outgrowth assays using ELISA and confocal microscopy to determine the effect of increasing concentration of SCAP on the total neurite outgrowth and axonal targeting. In addition, we evaluated whether SCAP evoked axonal targeting in vivo using a matrigel subcutaneous implant assay. Data were analyzed by ANOVA with Bonferroni's post hoc test, and significance was set at p<0.05. The results demonstrated that SCAP release a soluble factor that regulates neurite outgrowth from cultured trigeminal neurons. Next, we demonstrated that this effect is completely abolished by pretreatment with a neutralizing antibody to brain-derived neurotrophic factor (BDNF), but not by antibodies to other neurotrophins. Further, SCAP release BDNF in a concentration-dependent manner as detected by ELISA, and trigger directed axonal targeting both in vitro and in vivo as demonstrated by microfluidic and matrigel implant experiments, respectively. Collectively, these results suggest that SCAP may be responsible for the chemical signal driving axons to target regenerated tissue via a BDNF-dependent mechanism.
Collapse
Affiliation(s)
- Jose Flavio A de Almeida
- 1 Department of Restorative Dentistry, Endodontics Division, Piracicaba Dental School, University of Campinas , Piracicaba, São Paulo, Brazil
| | | | | | | |
Collapse
|
46
|
Walocko FM, Khouri RK, Urbanchek MG, Levi B, Cederna PS. The potential roles for adipose tissue in peripheral nerve regeneration. Microsurgery 2015; 36:81-8. [PMID: 26773850 DOI: 10.1002/micr.22480] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 06/29/2015] [Accepted: 08/19/2015] [Indexed: 02/06/2023]
Abstract
INTRODUCTION This review summarizes current understanding about the role of adipose-derived tissues in peripheral nerve regeneration and discusses potential advances that would translate this approach into the clinic. METHODS We searched PubMed for in vivo, experimental studies on the regenerative effects of adipose-derived tissues on peripheral nerve injuries. We summarized the methods and results for the 42 experiments. RESULTS Adipose-derived tissues enhanced peripheral nerve regeneration in 86% of the experiments. Ninety-five percent evaluated purified, cultured, or differentiated adipose tissue. These approaches have regulatory and scaling burdens, restricting clinical usage. Only one experiment tested the ability of adipose tissue to enhance nerve regeneration in conjunction with nerve autografts, the clinical gold standard. CONCLUSION Scientific studies illustrate that adipose-derived tissues enhance regeneration of peripheral nerves. Before this approach achieves clinical acceptance, fat processing must become automated and regulatory approval achieved. Animal studies using whole fat grafts are greatly needed for clinical translation.
Collapse
Affiliation(s)
- Frances M Walocko
- Office of Medical Student Education, University of Michigan Medical School, Ann Arbor, MI.,Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Roger K Khouri
- Office of Medical Student Education, University of Michigan Medical School, Ann Arbor, MI.,Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Melanie G Urbanchek
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Benjamin Levi
- Office of Medical Student Education, University of Michigan Medical School, Ann Arbor, MI.,Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Paul S Cederna
- Office of Medical Student Education, University of Michigan Medical School, Ann Arbor, MI.,Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| |
Collapse
|
47
|
Impact of traumatic brain injury on sleep structure, electrocorticographic activity and transcriptome in mice. Brain Behav Immun 2015; 47:118-30. [PMID: 25576803 DOI: 10.1016/j.bbi.2014.12.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI), including mild TBI (mTBI), is importantly associated with vigilance and sleep complaints. Because sleep is required for learning, plasticity and recovery, we here evaluated the bidirectional relationship between mTBI and sleep with two specific objectives: (1) Test that mTBI rapidly impairs sleep-wake architecture and the dynamics of the electrophysiological marker of sleep homeostasis (i.e., non-rapid eye movement sleep delta (1-4Hz) activity); (2) evaluate the impact of sleep loss following mTBI on the expression of plasticity markers that have been linked to sleep homeostasis and on genome-wide gene expression. A closed-head injury model was used to perform a 48h electrocorticographic (ECoG) recording in mice submitted to mTBI or Sham surgery. mTBI was found to immediately decrease the capacity to sustain long bouts of wakefulness as well as the amplitude of the time course of ECoG delta activity during wakefulness. Significant changes in ECoG spectral activity during wakefulness, non-rapid eye movement and rapid eye movement sleep were observed mainly on the second recorded day. A second experiment was performed to measure gene expression in the cerebral cortex and hippocampus after a mTBI followed either by two consecutive days of 6h sleep deprivation (SD) or of undisturbed behavior (quantitative PCR and next-generation sequencing). mTBI modified the expression of genes involved in immunity, inflammation and glial function (e.g., chemokines, glial markers) and SD changed that of genes linked to circadian rhythms, synaptic activity/neuronal plasticity, neuroprotection and cell death and survival. SD appeared to affect gene expression in the cerebral cortex more importantly after mTBI than Sham surgery including that of the astrocytic marker Gfap, which was proposed as a marker of clinical outcome after TBI. Interestingly, SD impacted the hippocampal expression of the plasticity elements Arc and EfnA3 only after mTBI. Overall, our findings reveal alterations in spectral signature across all vigilance states in the first days after mTBI, and show that sleep loss post-mTBI reprograms the transcriptome in a brain area-specific manner and in a way that could be deleterious to brain recovery.
Collapse
|
48
|
Fang YL, Chen XG, W T G. Gene delivery in tissue engineering and regenerative medicine. J Biomed Mater Res B Appl Biomater 2014; 103:1679-99. [PMID: 25557560 DOI: 10.1002/jbm.b.33354] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/07/2014] [Accepted: 11/18/2014] [Indexed: 12/13/2022]
Abstract
As a promising strategy to aid or replace tissue/organ transplantation, gene delivery has been used for regenerative medicine applications to create or restore normal function at the cell and tissue levels. Gene delivery has been successfully performed ex vivo and in vivo in these applications. Excellent proliferation capabilities and differentiation potentials render certain cells as excellent candidates for ex vivo gene delivery for regenerative medicine applications, which is why multipotent and pluripotent cells have been intensely studied in this vein. In this review, gene delivery is discussed in detail, along with its applications to tissue engineering and regenerative medicine. A definition of a stem cell is compared to a definition of a stem property, and both provide the foundation for an in-depth look at gene delivery investigations from a germ lineage angle.
Collapse
Affiliation(s)
- Y L Fang
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| | - X G Chen
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| | - Godbey W T
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| |
Collapse
|
49
|
Farrand AQ, Gregory RA, Scofield MD, Helke KL, Boger HA. Effects of aging on glutamate neurotransmission in the substantia nigra of Gdnf heterozygous mice. Neurobiol Aging 2014; 36:1569-76. [PMID: 25577412 DOI: 10.1016/j.neurobiolaging.2014.11.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 11/11/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) helps protect dopaminergic neurons in the nigrostriatal tract. Although the cause of nigrostriatal degeneration is unknown, one theory is that excess glutamate from the subthalamic nucleus results in excitotoxic events in the substantia nigra (SN). Because dopaminergic degeneration is accompanied by a reduction in GDNF, we examined glutamate neurotransmission in the SN using a Gdnf heterozygous mouse model (Gdnf(+/-)) at 8 and 12 months of age. At 8 months, Gdnf(+/-) mice have greater glutamate release and higher basal glutamate levels, which precede the SN dopaminergic degeneration observed at 12 months of age. However, at 12 months, Gdnf(+/-) mice have lower basal levels of glutamate and less glutamate release than wild-type mice. Also at 8 months, Gdnf(+/-) mice have lower levels of glutamate transporter-1 and greater glial fibrillary acidic protein levels in the SN compared with wild-type mice, differences that increase with age. These data suggest that reduced levels of GDNF induce excess glutamate release and dysregulation of glutamate transporter-1, causing excitotoxicity in the SN that precedes dopaminergic degeneration.
Collapse
Affiliation(s)
- Ariana Q Farrand
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Rebecca A Gregory
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Michael D Scofield
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Kristi L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Heather A Boger
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
50
|
Abdanipour A, Schluesener HJ, Tiraihi T, Noori-Zadeh A. Systemic administration of valproic acid stimulates overexpression of microtubule-associated protein 2 in the spinal cord injury model to promote neurite outgrowth. Neurol Res 2014; 37:223-8. [DOI: 10.1179/1743132814y.0000000438] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|