1
|
Biancotti JC, Sescleifer AM, Sferra SR, Penikis AB, Halbert-Elliott KM, Bubb CR, Kunisaki SM. Maternal Minocycline as Fetal Therapy in a Rat Model of Myelomeningocele. J Surg Res 2024; 301:696-703. [PMID: 39168042 DOI: 10.1016/j.jss.2024.07.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION This study aimed to investigate whether the maternal administration of minocycline, a tetracycline antibiotic known to have anti-inflammatory and neuroprotective properties in models of neural injury, reduces inflammation and neural cell death in a fetal rat model of myelomeningocele (MMC). METHODS E10 pregnant rats were gavaged with olive oil or olive oil + retinoic acid to induce fetal MMC. At E12, the dams were exposed to regular drinking water or water containing minocycline (range, 40-140 mg/kg/day). At E21, fetal lumbosacral spinal cords were isolated for immunohistochemistry and quantitative gene expression studies focused on microglia activity, inflammation, and apoptosis (P < 0.05). RESULTS There was a trend toward decreased activated Iba1+ microglial cells within the dorsal spinal cord of MMC pups following minocycline exposure when compared to water (H2O) alone (P = 0.052). Prenatal minocycline exposure was correlated with significantly reduced expression of the proinflammatory cytokine, IL-6 (minocycline: 1.75 versus H2O: 3.52, P = 0.04) and apoptosis gene, Bax (minocycline: 0.71 versus H2O: 1.04, P < 0.001) among MMC pups. CONCLUSIONS This study found evidence that the maternal administration of minocycline reduces selected markers of inflammation and apoptosis within the exposed dorsal spinal cords of fetal MMC rats. Further study of minocycline as a novel prenatal treatment strategy to mitigate spinal cord damage in MMC is warranted.
Collapse
Affiliation(s)
- Juan C Biancotti
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Anne M Sescleifer
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shelby R Sferra
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Annalise B Penikis
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kyra M Halbert-Elliott
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ciaran R Bubb
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shaun M Kunisaki
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
2
|
Liu R, Meng X, Yu X, Wang G, Dong Z, Zhou Z, Qi M, Yu X, Ji T, Wang F. From 2D to 3D Co-Culture Systems: A Review of Co-Culture Models to Study the Neural Cells Interaction. Int J Mol Sci 2022; 23:13116. [PMID: 36361902 PMCID: PMC9656609 DOI: 10.3390/ijms232113116] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 06/11/2024] Open
Abstract
The central nervous system (CNS) controls and regulates the functional activities of the organ systems and maintains the unity between the body and the external environment. The advent of co-culture systems has made it possible to elucidate the interactions between neural cells in vitro and to reproduce complex neural circuits. Here, we classified the co-culture system as a two-dimensional (2D) co-culture system, a cell-based three-dimensional (3D) co-culture system, a tissue slice-based 3D co-culture system, an organoid-based 3D co-culture system, and a microfluidic platform-based 3D co-culture system. We provide an overview of these different co-culture models and their applications in the study of neural cell interaction. The application of co-culture systems in virus-infected CNS disease models is also discussed here. Finally, the direction of the co-culture system in future research is prospected.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiaoting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiyao Yu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Guoqiang Wang
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhiyong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhengjie Zhou
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Mingran Qi
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiao Yu
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Tong Ji
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
3
|
The potential use of tetracyclines in neurodegenerative diseases and the role of nano-based drug delivery systems. Eur J Pharm Sci 2022; 175:106237. [PMID: 35710076 DOI: 10.1016/j.ejps.2022.106237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/07/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022]
Abstract
Neurodegenerative diseases are still a challenge for effective treatments. The high cost of approved drugs, severity of side effects, injection site pain, and restrictions on drug delivery to the Central Nervous System (CNS) can overshadow the management of these diseases. Due to the chronic and progressive evolution of neurodegenerative disorders and since there is still no cure for them, new therapeutic strategies such as the combination of several drugs or the use of existing drugs with new therapeutic applications are valuable strategies. Tetracyclines are traditionally classified as antibiotics. However, in this class of drugs, doxycycline and minocycline exhibit also anti-inflammatory effects by inhibiting microglia/macrophages. Hence, they have been studied as potential agents for the treatment of neurodegenerative diseases. The results of in vitro and in vivo studies confirm the effective role of these two drugs as anti-inflammatory agents in experimentally induced models of neurodegenerative diseases. In clinical studies, satisfactory results have been obtained in Multiple sclerosis (MS) but not yet in other disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), or Amyotrophic lateral sclerosis (ALS). In recent years, researchers have developed and evaluated nanoparticulate drug delivery systems to improve the clinical efficacy of these two tetracyclines for their potential application in neurodegenerative diseases. This study reviews the neuroprotective roles of minocycline and doxycycline in four of the main neurodegenerative disorders: AD, PD, ALS and MS. Moreover, the potential applications of nanoparticulate delivery systems developed for both tetracyclines are also reviewed.
Collapse
|
4
|
Rathore RS, R Ayyannan S, Mahto SK. Emerging three-dimensional neuronal culture assays for neurotherapeutics drug discovery. Expert Opin Drug Discov 2022; 17:619-628. [DOI: 10.1080/17460441.2022.2061458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Rahul S Rathore
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| | - Senthil R Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| | - Sanjeev K Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| |
Collapse
|
5
|
Keilhoff G, Pinkernelle J, Fansa H. The Ryanodine receptor stabilizer S107 fails to support motor neuronal neuritogenesis in vitro. Tissue Cell 2021; 73:101625. [PMID: 34419737 DOI: 10.1016/j.tice.2021.101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/02/2021] [Accepted: 08/15/2021] [Indexed: 11/30/2022]
Abstract
Calcium homeostasis is essential for neuronal cell survival/differentiation. Imbalance of the Ca2+ homeostasis due to excessive Ca2+ overload is essential for spinal cord injury (SCI). The overload resulted from Ca2+ flux across the plasma membrane and from internal Ca2+ store release (mitochondria, endoplasmic reticulum, ER). Inositol trisphosphate receptors (IP3R) and ryanodine receptors (RyR) are involved in releasing Ca2+ from ER contributing to axonal degeneration following SCI. In turn, block of both receptors is axoprotective. The calstabin RyR subunit, stabilizing the channel in a state of reduced activity, prevents pathological Ca2+ release too. We investigated whether S107, a RyR-stabilizing compound (Rycal), is beneficial for survival and neuritogenesis of spinal cord motor neurons in vitro. We used a spinal cord slice model and the motor neuron-like NSC-34 cell line. Effects of S107 were tested by propidium iodide/fluorescein diacetate vital staining, mitotic index determination via BrdU-incorporation, and neurite sprouting parameters. Results showed that S107 (i) had no effect on gliosis resulting from slices preparation; (ii) had no effect on motor neuronal survival and proliferation; and (iii) impaired neurite sprouting, no matter whether it was a differentiation (NSC-34 cells) or regeneration (spinal cord slices) process. The results underline the need for a flexible Ca2+homeostasis provided by the ER for re-initiation of neuritogenesis.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, 39120, Magdeburg, Germany.
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, 39120, Magdeburg, Germany
| | - Hisham Fansa
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, OWL-University, 33604, Bielefeld, Germany; Department of Plastic Surgery, and Breast Centre, Spital Zollikerberg, 8125, Zollikerberg, Switzerland
| |
Collapse
|
6
|
Keilhoff G, Ludwig C, Pinkernelle J, Lucas B. Effects of Gynostemma pentaphyllum on spinal cord motor neurons and microglial cells in vitro. Acta Histochem 2021; 123:151759. [PMID: 34425524 DOI: 10.1016/j.acthis.2021.151759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 07/11/2021] [Indexed: 11/18/2022]
Abstract
The regenerative capability of spinal cord neurons is limited to impossible. Thus, experimental approaches supporting reconstruction/regeneration are in process. This study focused on the evaluation of the protective potency of an extract from Gynostemma pentaphyllum (GP), a plant used in traditional medicine with anti-oxidative and neuroprotective activities, in vitro on organotypic spinal cord cultures, the motor-neuron-like NSC-34 cell line and the microglial cell line BV-2. Organotypic cultures were mechanically stressed by the slicing procedure and the effect of GP on motor neuron survival and neurite sprouting was tested by immunohistochemistry. NSC-34 cells were neuronal differentiated by using special medium. Afterwards, cell survival (propidium iodide/fluorescein diacetate labeling), proliferation (BrdU-incorporation), and neurite sprouting were evaluated. BV-2 cells were stimulated with LPS/interferon γ and subjected to migration assay and nanoparticle uptake. Cell survival, proliferation and the expression pattern of different microglial activation markers (cFOS, iNOS) as well as transcription factors (PPARγ, YB1) were analyzed. In organotypic cultures, high-dose GP supported survival of motor neurons and especially of the neuronal fiber network. Despite reduced neurodegeneration, however, there was a GP-mediated activation of astro- and microglia. In NSC-34 cells, high-dosed GP had degenerative and anti-proliferative effects, but only in normal medium. Moreover, GP supported the neuro-differentiation ability. In BV-2 cells, high-dosed GP was toxic. In lower dosages, GP affected cell survival and proliferation when combined with LPS/interferon γ. Nanoparticle uptake, migration ability, and the transcription factor PPARγ, however, GP affected directly. The data suggest positive effects of GP on injured spinal motor neurons. Moreover, GP activated microglial cells. The dual role of microglia (protective/detrimental) in neurodegenerative processes required further experiments to enhance the knowledge about GP effects. Therefore, a possible clinical use of GP in spinal cord injuries is still a long way off.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany.
| | - Christina Ludwig
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| | - Benjamin Lucas
- Dept. of Trauma Surgery, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| |
Collapse
|
7
|
Afshari K, Momeni Roudsari N, Lashgari NA, Haddadi NS, Haj-Mirzaian A, Hassan Nejad M, Shafaroodi H, Ghasemi M, Dehpour AR, Abdolghaffari AH. Antibiotics with therapeutic effects on spinal cord injury: a review. Fundam Clin Pharmacol 2020; 35:277-304. [PMID: 33464681 DOI: 10.1111/fcp.12605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/06/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Accumulating evidence indicates that a considerable number of antibiotics exert anti-inflammatory and neuroprotective effects in different central and peripheral nervous system diseases including spinal cord injury (SCI). Both clinical and preclinical studies on SCI have found therapeutic effects of antibiotics from different families on SCI. These include macrolides, minocycline, β-lactams, and dapsone, all of which have been found to improve SCI sequels and complications. These antibiotics may target similar signaling pathways such as reducing inflammatory microglial activity, promoting autophagy, inhibiting neuronal apoptosis, and modulating the SCI-related mitochondrial dysfunction. In this review paper, we will discuss the mechanisms underlying therapeutic effects of these antibiotics on SCI, which not only could supply vital information for investigators but also guide clinicians to consider administering these antibiotics as part of a multimodal therapeutic approach for management of SCI and its complications.
Collapse
Affiliation(s)
- Khashayar Afshari
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.,Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran
| | - Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran
| | - Nazgol-Sadat Haddadi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.,Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Malihe Hassan Nejad
- Department of Infectious Diseases, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, 1419733141, Iran
| | - Hamed Shafaroodi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts School of Medicine, Worcester, MA, 01655, USA
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran.,Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, 31375-1369, Iran.,Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
| |
Collapse
|
8
|
Ashford BA, Boche D, Cooper-Knock J, Heath PR, Simpson JE, Highley JR. Review: Microglia in motor neuron disease. Neuropathol Appl Neurobiol 2020; 47:179-197. [PMID: 32594542 DOI: 10.1111/nan.12640] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023]
Abstract
Motor Neuron Disease (MND) is a fatal neurodegenerative condition, which is characterized by the selective loss of the upper and lower motor neurons. At the sites of motor neuron injury, accumulation of activated microglia, the primary immune cells of the central nervous system, is commonly observed in both human post mortem studies and animal models of MND. Microglial activation has been found to correlate with many clinical features and importantly, the speed of disease progression in humans. Both anti-inflammatory and pro-inflammatory microglial responses have been shown to influence disease progression in humans and models of MND. As such, microglia could both contribute to and protect against inflammatory mechanisms of pathogenesis in MND. While murine models have characterized the microglial response to MND, these studies have painted a complex and often contradictory picture, indicating a need for further characterization in humans. This review examines the potential role microglia play in MND in human and animal studies. Both the pro-inflammatory and anti-inflammatory responses will be addressed, throughout the course of disease, followed by the potential of microglia as a target in the development of disease-modifying treatments for MND.
Collapse
Affiliation(s)
| | - D Boche
- University of Southampton, Southampton, UK
| | | | - P R Heath
- University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
9
|
Puty B, Nogueira ICDC, Nogueira LS, Vasconcelos CP, Araújo TMC, Bittencourt LO, Ferreira RDO, Oliveira EHCD, Leal WG, Lima RR. Genotoxic effect of non-lethal concentrations of minocycline in human glial cell culture. Biomed Pharmacother 2020; 128:110285. [PMID: 32485569 DOI: 10.1016/j.biopha.2020.110285] [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: 03/21/2020] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 01/01/2023] Open
Abstract
Minocycline has been proposed as a neuroprotective agent with pleiotropic effects on several experimental models of neurodegenerative diseases, including microglial inhibition. However, although most studies have focused on the central actions of minocycline in affecting microglial functions, other central nervous system (CNS) cell types may also be affected by this drug toxicity. Hence, considering that glial cells play a pivotal role on CNS physiology and are the main responsible for neuronal integrity, a comprehensive investigation on the effects of minocycline treatment on human glial cells is mandatory before translational studies to afford neuroprotection in humans. Therefore, we explored the cytotoxic and genotoxic effects of minocycline at different concentrations in glial cells using an in vitro model. To achieve this, U87 glial cell were exposed to 10-50 μg/mL for 24 h. After exposure, cell viability, general metabolic status and genotoxic assays were performed. No changes were observed in cell viability, however, the general metabolic status decreased over 20 μg/mL. In addition, although no chromossome aberrations were observed, evidences of genotoxicity, such as increase on micronucleus, buds and bridges, were observed from 10 μg/mL. These results suggest that minocycline may induce genotoxic effects even at concentrations considered previously safe and should be used with caution in translational studies.
Collapse
Affiliation(s)
- Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
| | - Iago César da Costa Nogueira
- Laboratory of Cell Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Lygia S Nogueira
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
| | | | - Teka Mayara Corrêa Araújo
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
| | - Railson de Oliveira Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil
| | | | - Walace Gomes Leal
- Laboratory of Experimental Neuroprotection and Neuroregeneration, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Science, Federal University of Pará, Belém, Brazil.
| |
Collapse
|
10
|
CX3CR1-Targeted PLGA Nanoparticles Reduce Microglia Activation and Pain Behavior in Rats with Spinal Nerve Ligation. Int J Mol Sci 2020; 21:ijms21103469. [PMID: 32423102 PMCID: PMC7279022 DOI: 10.3390/ijms21103469] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Activation of CX3CR1 in microglia plays an important role in the development of neuropathic pain. Here, we investigated whether neuropathic pain could be attenuated in spinal nerve ligation (SNL)-induced rats by reducing microglial activation through the use of poly(D,L-lactic-co-glycolic acid) (PLGA)-encapsulated CX3CR1 small-interfering RNA (siRNA) nanoparticles. After confirming the efficacy and specificity of CX3CR1 siRNA, as evidenced by its anti-inflammatory effects in lipopolysaccharide-stimulated BV2 cells in vitro, PLGA-encapsulated CX3CR1 siRNA nanoparticles were synthesized by sonication using the conventional double emulsion (W/O/W) method and administered intrathecally into SNL rats. CX3CR1 siRNA-treated rats exhibited significant reductions in the activation of microglia in the spinal dorsal horn and a downregulation of proinflammatory mediators, as well as a significant attenuation of mechanical allodynia. These data indicate that the PLGA-encapsulated CX3CR1 siRNA nanoparticles effectively reduce neuropathic pain in SNL-induced rats by reducing microglial activity and the expression of proinflammatory mediators. Therefore, we believe that PLGA-encapsulated CX3CR1 siRNA nanoparticles represent a valuable new treatment option for neuropathic pain.
Collapse
|
11
|
Patar A, Dockery P, McMahon S, Howard L. Ex Vivo Rat Transected Spinal Cord Slices as a Model to Assess Lentiviral Vector Delivery of Neurotrophin-3 and Short Hairpin RNA against NG2. BIOLOGY 2020; 9:biology9030054. [PMID: 32183469 PMCID: PMC7150802 DOI: 10.3390/biology9030054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 01/06/2023]
Abstract
The failure of the spinal cord to regenerate can be attributed both to a lack of trophic support for regenerating axons and to upregulation of inhibitory factors such as chondroitin sulphate proteoglycans including NG2 following injury. Lentiviral vector-mediated gene therapy is a possible strategy for treating spinal cord injury (SCI). This study investigated the effect of lentiviral vectors expressing Neurotrophin-3 (NT-3) and short-hairpin RNA against NG2 (NG2 sh) to enhance neurite outgrowth in in vitro and ex vivo transection injury models. Conditioned medium from cells transduced with NT-3 or shNG2 lentiviruses caused a significant increase in neurite length of primary dorsal root ganglia neurons compared to the control group in vitro. In an ex vivo organotypic slice culture (OSC) transduction with Lenti-NT-3 promoted axonal growth. Transducing OSCs with a combination of Lenti-NT-3/NG2 sh lead to a further increase in axonal growth but only in injured slices and only within the region adjacent to the site of injury. These findings suggest that the combination of lentiviral NT-3 and NG2 sh reduced NG2 levels and provided a more favourable microenvironment for neuronal regeneration after SCI. This study also shows that OSCs may be a useful platform for studying glial scarring and potential SCI treatments.
Collapse
Affiliation(s)
- Azim Patar
- Discipline of Anatomy, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 YR71 Galway, Ireland; (A.P.); (P.D.)
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Gelugor 11800, Malaysia
| | - Peter Dockery
- Discipline of Anatomy, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 YR71 Galway, Ireland; (A.P.); (P.D.)
| | - Siobhan McMahon
- Discipline of Anatomy, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 YR71 Galway, Ireland; (A.P.); (P.D.)
- Correspondence: (S.M.); (L.H.); Tel.: +353-91495268 (L.H.)
| | - Linda Howard
- Regenerative Medicine Institute (REMEDI), College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 YR71 Galway, Ireland
- Correspondence: (S.M.); (L.H.); Tel.: +353-91495268 (L.H.)
| |
Collapse
|
12
|
He L, Xu R, Chen Y, Liu X, Pan Y, Cao S, Xu T, Tian H, Zeng J. Intra-CA1 Administration of Minocycline Alters the Expression of Inflammation-Related Genes in Hippocampus of CCI Rats. Front Mol Neurosci 2019; 12:248. [PMID: 31708740 PMCID: PMC6822549 DOI: 10.3389/fnmol.2019.00248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 09/26/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Li He
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Rui Xu
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Yuanshou Chen
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Xiaohong Liu
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Youfu Pan
- Department of Genetics, Zunyi Medical University, Zunyi, China
| | - Song Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical Univerisity, Zunyi, China
| | - Tao Xu
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Hong Tian
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Junwei Zeng
- Department of Physiology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
13
|
Shurin GV, Kruglov O, Ding F, Lin Y, Hao X, Keskinov AA, You Z, Lokshin AE, LaFramboise WA, Falo LD, Shurin MR, Bunimovich YL. Melanoma-Induced Reprogramming of Schwann Cell Signaling Aids Tumor Growth. Cancer Res 2019; 79:2736-2747. [PMID: 30914431 DOI: 10.1158/0008-5472.can-18-3872] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/29/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment has been compared with a nonhealing wound involving a complex interaction between multiple cell types. Schwann cells, the key regulators of peripheral nerve repair, have recently been shown to directly affect nonneural wound healing. Their role in cancer progression, however, has been largely limited to neuropathic pain and perineural invasion. In this study, we showed that melanoma activated otherwise dormant functions of Schwann cells aimed at nerve regeneration and wound healing. Such reprogramming of Schwann cells into repair-like cells occurred during the destruction and displacement of neurons as the tumor expanded and via direct signaling from melanoma cells to Schwann cells, resulting in activation of the nerve injury response. Melanoma-activated Schwann cells significantly altered the microenvironment through their modulation of the immune system and the extracellular matrix in a way that promoted melanoma growth in vitro and in vivo. Local inhibition of Schwann cell activity following cutaneous sensory nerve transection in melanoma orthotopic models significantly decreased the rate of tumor growth. Tumor-associated Schwann cells, therefore, can have a significant protumorigenic effect and may present a novel target for cancer therapy. SIGNIFICANCE: These findings reveal a role of the nerve injury response, particularly through functions of activated Schwann cells, in promoting melanoma growth.
Collapse
Affiliation(s)
- Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Oleg Kruglov
- Department of Dermatology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Fei Ding
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Yan Lin
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Xingxing Hao
- Department of Dermatology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anton A Keskinov
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Zhaoyang You
- Department of Dermatology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Hillman Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anna E Lokshin
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - William A LaFramboise
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Hillman Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Hillman Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Yuri L Bunimovich
- Department of Dermatology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania. .,Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
14
|
Patar A, Dockery P, Howard L, McMahon SS. Cell viability in three ex vivo rat models of spinal cord injury. J Anat 2018; 234:244-251. [PMID: 30417349 DOI: 10.1111/joa.12909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2018] [Indexed: 12/18/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating disorder that has a poor prognosis of recovery. Animal models of SCI are useful to understand the pathophysiology of SCI and the potential use of therapeutic strategies for human SCI. Ex vivo models of central nervous system (CNS) trauma, particularly mechanical trauma, have become important tools to complement in vivo models of injury in order to reproduce the sequelae of human CNS injury. Ex vivo organotypic slice cultures (OSCs) provide a reliable model platform for the study of cell dynamics and therapeutic intervention following SCI. In addition, these ex vivo models support the 3R concept of animal use in SCI research - replacement, reduction and refinement. Ex vivo models cannot be used to monitor functional recovery, nor do they have the intact blood supply of the in vivo model systems. However, the ex vivo models appear to reproduce many of the post traumatic events including acute and secondary injury mechanisms. Several well-established OSC models have been developed over the past few years for experimental spinal injuries ex vivo in order to understand the biological response to injury. In this study, we investigated cell viability in three ex vivo OSC models of SCI: stab injury, transection injury and contusion injury. Injury was inflicted in postnatal day 4 rat spinal cord slices. Stab injury was performed using a needle on transverse slices of spinal cord. Transection injury was performed on longitudinal slices of spinal cord using a double blade technique. Contusion injury was performed on longitudinal slices of spinal cord using an Infinite Horizon impactor device. At days 3 and 10 post-injury, viability was measured using dual staining for propidium iodide and fluorescein diacetate. In all ex vivo SCI models, the slices showed more live cells than dead cells over 10 days in culture, with higher cell viability in control slices compared with injured slices. Although no change in cell viability was observed between time-points in stab- and contusion-injured OSCs, a reduction in cell viability was observed over time in transection-injured OSCs. Taken together, ex vivo SCI models are a useful and reliable research tool that reduces the cost and time involved in carrying out animal studies. The use of OSC models provides a simple way to study the cellular consequences following SCI, and they can also be used to investigate potential therapeutics regimes for the treatment of SCI.
Collapse
Affiliation(s)
- Azim Patar
- Discipline of Anatomy and NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland.,Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Peter Dockery
- Discipline of Anatomy, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Linda Howard
- Regenerative Medicine Institute (REMEDI), College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Siobhan S McMahon
- Discipline of Anatomy and NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
15
|
Patar A, Dockery P, Howard L, McMahon S. Analysis of reactive astrocytes and NG2 proteoglycan in ex vivo rat models of spinal cord injury. J Neurosci Methods 2018; 311:418-425. [PMID: 30267723 DOI: 10.1016/j.jneumeth.2018.09.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND The use of animals to model spinal cord injury (SCI) requires extensive post-operative care and can be expensive, which makes an alternative model extremely attractive. The use ofex vivo slice cultures is an alternative way to study the pathophysiological changes that can mimic in vivo conditions and support the 3Rs (replacement, reduction and refinement) of animal use in SCI research models. NEW METHOD In this study the presence of reactive astrocytes and NG2 proteoglycans was investigated in two ex vivo models of SCI; stab injury and transection injury. Stereological analysis to measure immunohistochemical staining was performed on the scar and injury zones to detect astrocytes and the chondroitin sulphate proteoglycan NG2. RESULTS The volume fraction (Vv) of reactive astrocytes and NG2 proteoglycans increased significantly between day 3 and day 10 post injury in both ex vivo models. This data shows how ex vivo SCI models are a useful research tool allowing reduction of research cost and time involved in carrying out animal studies, as well as reducing the numbers of animals used. COMPARISON WITH EXISTING METHOD This is the first evidence of an ex vivo stab injury model of SCI and also the first comparison of immunohistochemical staining for injury markers within stab injured and transection injured ex vivo slice cultures. CONCLUSIONS The use of organotypic slice culture models provide a simple way to study the cellular consequences following SCI and they can also be used as a platform for potential therapeutics regimes for the treatment of SCI.
Collapse
Affiliation(s)
- Azim Patar
- Discipline of Anatomy and NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland; Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Malaysia
| | - Peter Dockery
- Discipline of Anatomy, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| | - Linda Howard
- Regenerative Medicine Institute (REMEDI), College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| | - Siobhan McMahon
- Discipline of Anatomy and NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland.
| |
Collapse
|
16
|
Drenger B, Blanck TJJ, Piskoun B, Jaffrey E, Recio-Pinto E, Sideris A. Minocycline Before Aortic Occlusion Reduces Hindlimb Motor Impairment, Attenuates Spinal Cord Damage and Spinal Astrocytosis, and Preserve Neuronal Cytoarchitecture in the Rat. J Cardiothorac Vasc Anesth 2018; 33:1003-1011. [PMID: 30195965 DOI: 10.1053/j.jvca.2018.07.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Indexed: 11/11/2022]
Abstract
OBJECTIVES Spinal cord ischemia secondary to trauma or a vascular occlusive event is a threatening phenomenon. The neuroprotective properties of minocycline have been shown in several models of central nervous system diseases and after spinal cord ischemia; however, the benefit of using the drug requires additional confirmation in different animal models. Astrocytes are essential as regulators of neuronal functions and for providing nutrients. The authors hypothesized that astrocytes in the spinal cord may be an important target for minocycline action after ischemia and thus in the prevention of secondary spreading damage. DESIGN A prospective, randomized animal study. SETTING University research laboratory, single institution. PARTICIPANTS Adult male Sprague Dawley rats, weighing between 400 and 450 g. INTERVENTIONS A model of spinal cord ischemia in the rat was used for this study to determine whether a single, high-dose (10 mg/kg) of minocycline protects against damage to the neuronal cytoskeleton, both in the white and gray matter, and whether it reduces glial fibrillary acidic protein levels, which is an index for prevention of astrocyte activation during ischemia. Thirty minutes before thoracic aorta occlusion, minocycline was administered for 18 minutes using a 2 F Fogarty catheter. MEASUREMENTS AND MAIN RESULTS Minocycline given prophylactically significantly mitigated severe hindlimb motor impairment and reduced glial fibrillary acidic protein plus astrocytosis in both the white and gray matter of the spinal cord, caudal to the occlusion. Neuronal histologic cytoarchitecture, which was severely and significantly compromised in control animals, was preserved in the minocycline-treated animals. CONCLUSIONS This study's data imply that minocycline may attenuate reactive astrocytosis in response to injury with better neurologic outcome in a model of spinal cord ischemia in rats. The data suggest that future use of minocycline, clinically, might be advantageous in surgeries with a potential risk for paraplegia due to spinal cord ischemia.
Collapse
Affiliation(s)
- Benjamin Drenger
- Department of Anesthesiology and Critical Care Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | - Thomas J J Blanck
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Medical Center, New York, NY
| | - Boris Piskoun
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Medical Center, New York, NY
| | - E Jaffrey
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Medical Center, New York, NY
| | - Esperanza Recio-Pinto
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Medical Center, New York, NY; Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY
| | - Alexandra Sideris
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Medical Center, New York, NY; Department of Perioperative Care and Pain Medicine, New York University Langone Medical Center, New York, NY
| |
Collapse
|
17
|
Vodret S, Bortolussi G, Iaconcig A, Martinelli E, Tiribelli C, Muro AF. Attenuation of neuro-inflammation improves survival and neurodegeneration in a mouse model of severe neonatal hyperbilirubinemia. Brain Behav Immun 2018; 70:166-178. [PMID: 29458193 DOI: 10.1016/j.bbi.2018.02.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/06/2018] [Accepted: 02/15/2018] [Indexed: 01/21/2023] Open
Abstract
All pre-term newborns and a high proportion of term newborns develop neonatal jaundice. Neonatal jaundice is usually a benign condition and self-resolves within few days after birth. However, a combination of unfavorable complications may lead to acute hyperbilirubinemia. Excessive hyperbilirubinemia may be toxic for the developing nervous system leading to severe neurological damage and death by kernicterus. Survivors show irreversible neurological deficits such as motor, sensitive and cognitive abnormalities. Current therapies rely on the use of phototherapy and, in unresponsive cases, exchange transfusion, which is performed only in specialized centers. During bilirubin-induced neurotoxicity different molecular pathways are activated, ranging from oxidative stress to endoplasmic reticulum (ER) stress response and inflammation, but the contribution of each pathway in the development of the disease still requires further investigation. Thus, to increase our understanding of the pathophysiology of bilirubin neurotoxicity, encephalopathy and kernicterus, we pharmacologically modulated neurodegeneration and neuroinflammation in a lethal mouse model of neonatal hyperbilirubinemia. Treatment of mutant mice with minocycline, a second-generation tetracycline with anti-inflammatory and neuroprotective properties, resulted in a dose-dependent rescue of lethality, due to reduction of neurodegeneration and neuroinflammation, without affecting plasma bilirubin levels. In particular, rescued mice showed normal motor-coordination capabilities and behavior, as determined by the accelerating rotarod and open field tests, respectively. From the molecular point of view, rescued mice showed a dose-dependent reduction in apoptosis of cerebellar neurons and improvement of dendritic arborization of Purkinje cells. Moreover, we observed a decrease of bilirubin-induced M1 microglia activation at the sites of damage with a reduction in oxidative and ER stress markers in these cells. Collectively, these data indicate that neurodegeneration and neuro-inflammation are key factors of bilirubin-induced neonatal lethality and neuro-behavioral abnormalities. We propose that the application of pharmacological treatments having anti-inflammatory and neuroprotective effects, to be used in combination with the current treatments, may significantly improve the management of acute neonatal hyperbilirubinemia, protecting from bilirubin-induced neurological damage and death.
Collapse
Affiliation(s)
- Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
| | - Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy.
| | - Alessandra Iaconcig
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
| | - Elena Martinelli
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
| | - Claudio Tiribelli
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza, Trieste, Italy
| | - Andrés F Muro
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy.
| |
Collapse
|
18
|
Minocycline improves cardiac function after myocardial infarction in rats by inhibiting activation of PARP-1. Biomed Pharmacother 2018; 97:1119-1124. [DOI: 10.1016/j.biopha.2017.10.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 11/21/2022] Open
|
19
|
Chin TY, Kiat SS, Faizul HG, Wu W, Abdullah JM. The Effects of Minocycline on Spinal Root Avulsion Injury in Rat Model. Malays J Med Sci 2017; 24:31-39. [PMID: 28381927 DOI: 10.21315/mjms2017.24.1.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/12/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The neuroprotective role of minocycline in the treatment of brachial plexus injury is controversial. OBJECTIVE To study the neuroprotective effect of minocycline via different routes in adult Sprague Dawley rats with brachial plexus injury. METHODS The C7 nerve roots of the animals were avulsed via an anterior extravertebral approach. Traction force was used to transect the ventral motor nerve roots at the preganglionic level. Intraperitoneal and intrathecal minocycline (50 mg/kg for the first week and 25 mg/kg for the second week) were administered to promote motor healing. The spinal cord was harvested six weeks after the injury, and structural changes following the avulsion injury and pharmacological intervention were analysed. RESULTS Motor neuron death and microglial proliferation were observed after the administration of minocycline via two different routes (intraperitoneal and intrathecal) following traumatic avulsion injury of the ventral nerve root. The administration of intraperitoneal minocycline reduced the microglia count but increased the motor neuron count. Intrathecal minocycline also reduced the microglial count, with a greater reduction than in the intraperitoneal group, but it decreased the motor neuron count. CONCLUSIONS Intraperitoneal minocycline increased motor neuron survival by inhibiting microglial proliferation following traumatic avulsion injury of the nerve root. The inhibitory effect was augmented by the use of intrathecal minocycline, in which the targeted drug delivery method increased the bioavailability of the therapeutic agent. However, motor neuron survival was impaired at a higher concentration of minocycline via the intrathecal route due to the more efficient method of drug delivery. Microglial suppression via minocycline can have both beneficial and damaging effects, with a moderate dose being beneficial as regards motor neuron survival but a higher dose proving neurotoxic due to impairment of the glial response and Wallerian degeneration, which is a pre-requisite for regeneration.
Collapse
Affiliation(s)
- Tan Yew Chin
- Department of Neurosciences School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Center for Neuroscience Services and Research, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Sim Sze Kiat
- Department of Neurosciences School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, 94300 Kota Samarahan, Sarawak, Malaysia
| | - Hizal Ghazali Faizul
- Department of Neurosciences School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Wutian Wu
- Department of Anatomy, Hong Kong University, 21 Sassoon Road, Hong Kong
| | - Jafri Malin Abdullah
- Department of Neurosciences School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Center for Neuroscience Services and Research, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
20
|
Park HW, Jeon HJ, Chang MS. Vascular endothelial growth factor enhances axonal outgrowth in organotypic spinal cord slices via vascular endothelial growth factor receptor 1 and 2. Tissue Eng Regen Med 2016; 13:601-609. [PMID: 30603441 DOI: 10.1007/s13770-016-0051-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/13/2016] [Accepted: 06/01/2016] [Indexed: 01/26/2023] Open
Abstract
Enhancing adult nerve regeneration is a potential therapeutic strategy for treating spinal cord injury. Vascular endothelial growth factor (VEGF) is a major contributor to angiogenesis, which can reduce the spinal cord injury by inhibiting the inflammation and improve recovery after spinal cord injury. We have previously demonstrated that exogenous VEGF has neurotrophic effects on injured spinal nerves in organotypic spinal cord slice cultures. However, the mechanisms underlying the neurite growth by exogenous VEGF remain to be explored in spinal cord. In this study, we found out that exogenous VEGF mediated axonal outgrowth through VEGF receptor 1 (VEGFR1) and VEGFR2, both of which were expressed on organotypic spinal cord slices. Although VEGFR1 and VEGFR2 were constitutively expressed in some cells of control spinal cord slices, VEGF treatment upregulated expression of VEGFR1 and VEGFR2. Both VEGFR1 and VEGFR2 were expressed in neuronal cells as well as glial cells of organotypic spinal cord slices. We also observed that VEGF-induced axonal outgrowth was attenuated by a specific mitogen-activated protein kinase (MAPK) inhibitor PD98059 and a specific phosphoinositide 3-kinase (PI3K) inhibitor wortmannin. Thus, these findings suggest that these MAPK and PI3K pathways have important roles in regulating VEGF-induced axonal outgrowth in the postnatal spinal cord.
Collapse
Affiliation(s)
- Hwan-Woo Park
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 2Department of Cell Biology, College of Medicine, Konyang University, Daejeon, Korea
| | - Hyo-Jin Jeon
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Mi-Sook Chang
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 3Neuroscience Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
21
|
KEILHOFF GERBURG, LUCAS BENJAMIN, UHDE KATJA, FANSA HISHAM. Selected gene profiles of stressed NSC-34 cells and rat spinal cord following peripheral nerve reconstruction and minocycline treatment. Exp Ther Med 2016; 11:1685-1699. [PMID: 27168790 PMCID: PMC4840837 DOI: 10.3892/etm.2016.3130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/17/2015] [Indexed: 12/20/2022] Open
Abstract
The present study was conducted to investigate the effects of minocycline on the expression of selected transcriptional and translational profiles in the rat spinal cord following sciatic nerve (SNR) transection and microsurgical coaptation. The mRNA and protein expression levels of B cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), caspase-3, major histocompatibility complex I (MHC I), tumor necrosis factor-α (TNF-α), activating transcription factor 3 (ATF3), vascular endothelial growth factor (VEGF), matrix metalloproteinase 9 (MMP9), and growth associated protein-43 (GAP-43) were monitored in the rat lumbar spinal cord following microsurgical reconstruction of the sciatic nerves and minocycline treatment. The present study used semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. As a PCR analysis of spinal cord tissue enabled the examination of the expression patterns of all cell types including glia, the motorneuron-like NSC-34 cell line was used to investigate expression level changes in motorneurons. As stressors, oxygen glucose deprivation (OGD) and lipopolysaccharide (LPS) treatment were performed. SNR did not induce significant degeneration of ventral horn motorneurons, whereas microglia activation and synaptic terminal retraction were detectable. All genes were constitutively expressed at the mRNA and protein levels in untreated spinal cord and control cells. SNR significantly increased the mRNA expression levels of all genes, albeit only temporarily. In all genes except MMP9 and GAP-43, the induction was seen ipsilaterally and contralaterally. The effects of minocycline were moderate. The expression levels of MMP9, TNF-α, MHC I, VEGF, and GAP-43 were reduced, whereas those of Bax and Bcl-2 were unaffected. OGD, but not LPS, was toxic for NSC-34 cells. No changes in the expression levels of Bax, caspase-3, MHC I or ATF3 were observed. These results indicated that motorneurons were not preferentially or solely responsible for SNR-mediated upregulation of these genes. MMP9, TNF-α, VEGF and Bcl-2 were stress-activated. These results suggest that a substantial participation of motorneurons in gene expression levels in vivo. Minocycline was also shown to have inhibitory effects. The nuclear factor-κB signalling pathway may be a possible target of minocycline.
Collapse
Affiliation(s)
- GERBURG KEILHOFF
- Institute of Biochemistry and Cell Biology, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
| | - BENJAMIN LUCAS
- Institute of Biochemistry and Cell Biology, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
- Department of Trauma Surgery, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
| | - KATJA UHDE
- Institute of Biochemistry and Cell Biology, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
| | - HISHAM FANSA
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, Bielefeld D-33604, Germany
| |
Collapse
|
22
|
Geuna S, Raimondo S, Fregnan F, Haastert-Talini K, Grothe C. In vitromodels for peripheral nerve regeneration. Eur J Neurosci 2015; 43:287-96. [DOI: 10.1111/ejn.13054] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/03/2015] [Accepted: 08/20/2015] [Indexed: 01/10/2023]
Affiliation(s)
- S. Geuna
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute; University of Turin; Ospedale San Luigi, Regione Gonzole 10 10043 Orbassano Turin Italy
| | - S. Raimondo
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute; University of Turin; Ospedale San Luigi, Regione Gonzole 10 10043 Orbassano Turin Italy
| | - F. Fregnan
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute; University of Turin; Ospedale San Luigi, Regione Gonzole 10 10043 Orbassano Turin Italy
| | - K. Haastert-Talini
- Institute of Neuroanatomy; Hannover Medical School and Center for Systems Neuroscience (ZSN); Hannover Germany
| | - C. Grothe
- Institute of Neuroanatomy; Hannover Medical School and Center for Systems Neuroscience (ZSN); Hannover Germany
| |
Collapse
|
23
|
Pinkernelle J, Raffa V, Calatayud MP, Goya GF, Riggio C, Keilhoff G. Growth factor choice is critical for successful functionalization of nanoparticles. Front Neurosci 2015; 9:305. [PMID: 26388717 PMCID: PMC4557102 DOI: 10.3389/fnins.2015.00305] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/12/2015] [Indexed: 12/16/2022] Open
Abstract
Nanoparticles (NPs) show new characteristics compared to the corresponding bulk material. These nanoscale properties make them interesting for various applications in biomedicine and life sciences. One field of application is the use of magnetic NPs to support regeneration in the nervous system. Drug delivery requires a functionalization of NPs with bio-functional molecules. In our study, we functionalized self-made PEI-coated iron oxide NPs with nerve growth factor (NGF) and glial cell-line derived neurotrophic factor (GDNF). Next, we tested the bio-functionality of NGF in a rat pheochromocytoma cell line (PC12) and the bio-functionality of GDNF in an organotypic spinal cord culture. Covalent binding of NGF to PEI-NPs impaired bio-functionality of NGF, but non-covalent approach differentiated PC12 cells reliably. Non-covalent binding of GDNF showed a satisfying bio-functionality of GDNF:PEI-NPs, but turned out to be unstable in conjugation to the PEI-NPs. Taken together, our study showed the importance of assessing bio-functionality and binding stability of functionalized growth factors using proper biological models. It also shows that successful functionalization of magnetic NPs with growth factors is dependent on the used binding chemistry and that it is hardly predictable. For use as therapeutics, functionalization strategies have to be reproducible and future studies are needed.
Collapse
Affiliation(s)
- Josephine Pinkernelle
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
- Institute for Biochemistry and Cell Biology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| | - Vittoria Raffa
- Department of Biology, University of PisaPisa, Italy
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | | | - Gerado F. Goya
- Aragon Institute of Nanosciences, University of ZaragozaZaragoza, Spain
- Department of Condensed Matter Physics, University of ZaragozaSpain
| | - Cristina Riggio
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | - Gerburg Keilhoff
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| |
Collapse
|
24
|
Pandamooz S, Nabiuni M, Miyan J, Ahmadiani A, Dargahi L. Organotypic Spinal Cord Culture: a Proper Platform for the Functional Screening. Mol Neurobiol 2015; 53:4659-74. [PMID: 26310972 DOI: 10.1007/s12035-015-9403-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Recent improvements in organotypic slice culturing and its accompanying technological innovations have made this biological preparation increasingly useful ex vivo experimental model. Among organotypic slice cultures obtained from various central nervous regions, spinal cord slice culture is an absorbing model that represents several unique advantages over other current in vitro and in vivo models. The culture of developing spinal cord slices, as allows real-time observation of embryonic cells behaviors, is an instrumental platform for developmental investigation. Importantly, due to the ability of ex vivo models to recapitulate different aspects of corresponding in vivo conditions, these models have been subject of various manipulations to derive disease-relevant slice models. Moreover spinal cord slice cultures represent a potential platform for screening of different pharmacological agents and evaluation of cell transplantation and neuroregenerative materials. In this review, we will focus on studies carried out using the ex vivo model of spinal cord slice cultures and main advantages linked to practicality of these slices in both normal and neuropathological diseases and summarize them in different categories based on application.
Collapse
Affiliation(s)
- Sareh Pandamooz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Nabiuni
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Jaleel Miyan
- Neurobiology Research Group, Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Microglial Activation Promotes Cell Survival in Organotypic Cultures of Postnatal Mouse Retinal Explants. PLoS One 2015; 10:e0135238. [PMID: 26252475 PMCID: PMC4529135 DOI: 10.1371/journal.pone.0135238] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/20/2015] [Indexed: 02/03/2023] Open
Abstract
The role of microglia during neurodegeneration remains controversial. We investigated whether microglial cells have a neurotoxic or neuroprotective function in the retina. Retinal explants from 10-day-old mice were treated in vitro with minocycline to inhibit microglial activation, with LPS to increase microglial activation, or with liposomes loaded with clodronate (Lip-Clo) to deplete microglial cells. Flow cytometry was used to assess the viability of retinal cells in the explants and the TUNEL method to show the distribution of dead cells. The immunophenotypic and morphological features of microglia and their distribution were analyzed with flow cytometry and immunocytochemistry. Treatment of retinal explants with minocycline reduced microglial activation and simultaneously significantly decreased cell viability and increased the presence of TUNEL-labeled cell profiles. This treatment also prevented the migration of microglial cells towards the outer nuclear layer, where cell death was most abundant. The LPS treatment increased microglial activation but had no effect on cell viability or microglial distribution. Finally, partial microglial removal with Lip-Clo diminished the cell viability in the retinal explants, showing a similar effect to that of minocycline. Hence, cell viability is diminished in retinal explants cultured in vitro when microglial cells are removed or their activation is inhibited, indicating a neurotrophic role for microglia in this system.
Collapse
|
26
|
Nakano Y, Furube E, Morita S, Wanaka A, Nakashima T, Miyata S. Astrocytic TLR4 expression and LPS-induced nuclear translocation of STAT3 in the sensory circumventricular organs of adult mouse brain. J Neuroimmunol 2015; 278:144-58. [DOI: 10.1016/j.jneuroim.2014.12.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/13/2014] [Accepted: 12/13/2014] [Indexed: 12/26/2022]
|
27
|
Ali S, Driscoll HE, Newton VL, Gardiner NJ. Matrix metalloproteinase-2 is downregulated in sciatic nerve by streptozotocin induced diabetes and/or treatment with minocycline: Implications for nerve regeneration. Exp Neurol 2014; 261:654-65. [PMID: 25158309 PMCID: PMC4199570 DOI: 10.1016/j.expneurol.2014.08.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/07/2014] [Accepted: 08/11/2014] [Indexed: 12/13/2022]
Abstract
Minocycline is an inhibitor of matrix metalloproteinases (MMPs) and has been shown to have analgesic effects. Whilst increased expression of MMPs is associated with neuropathic pain, MMPs also play crucial roles in Wallerian degeneration and nerve regeneration. In this study we examined the expression of MMP-2, MMP-9 and tissue inhibitor of metalloproteinase (TIMP)-1/-2 in the sciatic nerve of control and streptozotocin-induced diabetic rats treated with either vehicle or minocycline by quantitative PCR and gelatin zymography. We assessed the effects of minocycline on nerve conduction velocity and intraepidermal nerve fibre (IENF) deficits in diabetic neuropathy and investigated the effects of minocycline or MMP-2 on neurite outgrowth from primary cultures of dissociated adult rat sensory neurons. We show that MMP-2 is expressed constitutively in the sciatic nerve in vivo and treatment with minocycline or diabetes leads to downregulation of MMP-2 expression and activity. The functional consequence of this is IENF deficits in minocycline-treated nondiabetic rats and an unsupportive microenvironment for regeneration in diabetes. Minocycline reduces levels of MMP-2 mRNA and nerve growth factor-induced neurite outgrowth. Furthermore, in vivo minocycline treatment reduces preconditioning-induced in vitro neurite outgrowth following a sciatic nerve crush. In contrast, the addition of active MMP-2 facilitates neurite outgrowth in the absence of neurotrophic support and pre-treatment of diabetic sciatic nerve substrata with active MMP-2 promotes a permissive environment for neurite outgrowth. In conclusion we suggest that MMP-2 downregulation may contribute to the regenerative deficits in diabetes. Minocycline treatment also downregulates MMP-2 activity and is associated with inhibitory effects on sensory neurons. Thus, caution should be exhibited with its use as the balance between beneficial and detrimental outcomes may be critical in assessing the benefits of using minocycline to treat diabetic neuropathy. MMP-2, but not MMP-9, is constitutively expressed in the adult rat sciatic nerve. Levels of cleaved active MMP-2 are reduced in sciatic nerve of diabetic rats. Active MMP-2 potentiates neurite outgrowth from sensory neurons. Minocycline reduces levels of MMP-2 mRNA and impairs NGF-induced neurite growth. Minocycline did not prevent nerve dysfunction in experimental diabetic neuropathy.
Collapse
Affiliation(s)
- Sumia Ali
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Heather E Driscoll
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Victoria L Newton
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Natalie J Gardiner
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
28
|
Mika J, Popiolek-Barczyk K, Rojewska E, Makuch W, Starowicz K, Przewlocka B. Delta-opioid receptor analgesia is independent of microglial activation in a rat model of neuropathic pain. PLoS One 2014; 9:e104420. [PMID: 25105291 PMCID: PMC4126741 DOI: 10.1371/journal.pone.0104420] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/09/2014] [Indexed: 12/15/2022] Open
Abstract
The analgesic effect of delta-opioid receptor (DOR) ligands in neuropathic pain is not diminished in contrast to other opioid receptor ligands, which lose their effectiveness as analgesics. In this study, we examine whether this effect is related to nerve injury-induced microglial activation. We therefore investigated the influence of minocycline-induced inhibition of microglial activation on the analgesic effects of opioid receptor agonists: morphine, DAMGO, U50,488H, DPDPE, Deltorphin II and SNC80 after chronic constriction injury (CCI) to the sciatic nerve in rats. Pre-emptive and repeated administration of minocycline (30 mg/kg, i.p.) over 7 days significantly reduced allodynia and hyperalgesia as measured on day 7 after CCI. The antiallodynic and antihyperalgesic effects of intrathecally (i.t.) administered morphine (10–20 µg), DAMGO (1–2 µg) and U50,488H (25–50 µg) were significantly potentiated in rats after minocycline, but no such changes were observed after DPDPE (10–20 µg), deltorphin II (1.5–15 µg) and SNC80 (10–20 µg) administration. Additionally, nerve injury-induced down-regulation of all types of opioid receptors in the spinal cord and dorsal root ganglia was not influenced by minocycline, which indicates that the effects of opioid ligands are dependent on other changes, presumably neuroimmune interactions. Our study of rat primary microglial cell culture using qRT-PCR, Western blotting and immunocytochemistry confirmed the presence of mu-opioid receptors (MOR) and kappa-opioid receptors (KOR), further we provide the first evidence for the lack of DOR on microglial cells. In summary, DOR analgesia is different from analgesia induced by MOR and KOR receptors because it does not dependent on injury-induced microglial activation. DOR agonists appear to be the best candidates for new drugs to treat neuropathic pain.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/therapeutic use
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/therapeutic use
- Animals
- Anti-Bacterial Agents/administration & dosage
- Anti-Bacterial Agents/therapeutic use
- Cells, Cultured
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use
- Gene Expression Regulation/drug effects
- Male
- Microglia/cytology
- Microglia/drug effects
- Microglia/metabolism
- Minocycline/administration & dosage
- Minocycline/therapeutic use
- Morphine/administration & dosage
- Morphine/therapeutic use
- Neuralgia/drug therapy
- Rats, Wistar
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
Collapse
Affiliation(s)
- Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- * E-mail: (BP); (JM)
| | | | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Starowicz
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- * E-mail: (BP); (JM)
| |
Collapse
|
29
|
Keilhoff G, Lucas B, Pinkernelle J, Steiner M, Fansa H. Effects of cerebrolysin on motor-neuron-like NSC-34 cells. Exp Cell Res 2014; 327:234-55. [PMID: 24997385 DOI: 10.1016/j.yexcr.2014.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/12/2014] [Accepted: 06/26/2014] [Indexed: 01/01/2023]
Abstract
Although the peripheral nervous system is capable of regeneration, this capability is limited. As a potential means of augmenting nerve regeneration, the effects of cerebrolysin (CL)--a proteolytic peptide fraction--were tested in vitro on the motor-neuron-like NSC-34 cell line and organotypic spinal cord cultures. Therefore, NSC-34 cells were subjected to mechanical stress by changing media and metabolic stress by oxygen glucose deprivation. Afterwards, cell survival/proliferation using MTT and BrdU-labeling (FACS) and neurite sprouting using ImageJ analysis were evaluated. Calpain-1, Src and α-spectrin protein expression were analyzed by Western blot. In organotypic cultures, the effect of CL on motor neuron survival and neurite sprouting was tested by immunohistochemistry. CL had a temporary anti-proliferative but initially neuroprotective effect on OGD-stressed NSC-34 cells. High-dosed or repeatedly applied CL was deleterious for cell survival. CL amplified neurite reconstruction to limited extent, affected calpain-1 protein expression and influenced calpain-mediated spectrin cleavage as a function of Src expression. In organotypic spinal cord slice cultures, CL was not able to support motor neuron survival/neurite sprouting. Moreover, it hampered astroglia and microglia activities. The data suggest that CL may have only isolated positive effects on injured spinal motor neurons. High-dosed or accumulated CL seemed to have adverse effects in treatment of spinal cord injury. Further experiments are required to optimize the conditions for a safe clinical administration of CL in spinal cord injuries.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Benjamin Lucas
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Michael Steiner
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Hisham Fansa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, Teutoburger Str. 50, D-33604 Bielefeld, Germany
| |
Collapse
|