1
|
Marzoog BA. Autophagy as an Anti-senescent in Aging Neurocytes. Curr Mol Med 2024; 24:182-190. [PMID: 36683318 DOI: 10.2174/1566524023666230120102718] [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: 07/31/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 01/24/2023]
Abstract
Neuron homeostasis is crucial for the organism, and its maintenance is multifactorial, including autophagy. The turnover of aberrant intracellular components is a fundamental pathogenetic mechanism for cell aging. Autophagy is involved in the acceleration of the neurocyte aging process and the modification of cell longevity. Neurocyte aging is a process of loss of cell identity through cellular and subcellular changes that include molecular loss of epigenetics, transcriptomic, proteomic, and autophagy dysfunction. Autophagy dysfunction is the hallmark of neurocyte aging. Cell aging is the credential feature of neurodegenerative diseases. Pathophysiologically, aged neurocytes are characterized by dysregulated autophagy and subsequently neurocyte metabolic stress, resulting in accelerated neurocyte aging. In particular, chaperone- mediated autophagy perturbation results in upregulated expression of aging and apoptosis genes. Aged neurocytes are also characterized by the down-regulation of autophagy-related genes, such as ATG5-ATG12, LC3-II / LC3-I ratio, Beclin-1, and p62. Slowing aging through autophagy targeting is sufficient to improve prognosis in neurodegenerative diseases. Three primary anti-senescent molecules are involved in the aging process: mTOR, AMPK, and Sirtuins. Autophagy therapeutic effects can be applied to reverse and slow aging. This article discusses current advances in the role of autophagy in neurocyte homeostasis, aging, and potential therapeutic strategies to reduce aging and increase cell longevity.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- National Research Mordovia State University, Bolshevitskaya Street, 68, Saransk, 430005, Rep. Mordovia, Russia
| |
Collapse
|
2
|
Koyama Y, Harada S, Sato T, Kobayashi Y, Yanagawa H, Iwahashi T, Tanaka H, Ohata K, Imai T, Ohta Y, Kamakura T, Kobayashi H, Inohara H, Shimada S. Therapeutic strategy for facial paralysis based on the combined application of Si-based agent and methylcobalamin. Biochem Biophys Rep 2022; 32:101388. [DOI: 10.1016/j.bbrep.2022.101388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/02/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022] Open
|
3
|
Lee JI, Park JH, Kim YR, Gwon K, Hwang HW, Jung G, Lee JY, Sun JY, Park JW, Shin JH, Ok MR. Delivery of nitric oxide-releasing silica nanoparticles for in vivo revascularization and functional recovery after acute peripheral nerve crush injury. Neural Regen Res 2022; 17:2043-2049. [PMID: 35142695 PMCID: PMC8848604 DOI: 10.4103/1673-5374.335160] [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] [Indexed: 11/19/2022] Open
Abstract
Nitric oxide (NO) has been shown to promote revascularization and nerve regeneration after peripheral nerve injury. However, in vivo application of NO remains challenging due to the lack of stable carrier materials capable of storing large amounts of NO molecules and releasing them on a clinically meaningful time scale. Recently, a silica nanoparticle system capable of reversible NO storage and release at a controlled and sustained rate was introduced. In this study, NO-releasing silica nanoparticles (NO-SNs) were delivered to the peripheral nerves in rats after acute crush injury, mixed with natural hydrogel, to ensure the effective application of NO to the lesion. Microangiography using a polymer dye and immunohistochemical staining for the detection of CD34 (a marker for revascularization) results showed that NO-releasing silica nanoparticles increased revascularization at the crush site of the sciatic nerve. The sciatic functional index revealed that there was a significant improvement in sciatic nerve function in NO-treated animals. Histological and anatomical analyses showed that the number of myelinated axons in the crushed sciatic nerve and wet muscle weight excised from NO-treated rats were increased. Moreover, muscle function recovery was improved in rats treated with NO-SNs. Taken together, our results suggest that NO delivered to the injured sciatic nerve triggers enhanced revascularization at the lesion in the early phase after crushing injury, thereby promoting axonal regeneration and improving functional recovery.
Collapse
Affiliation(s)
- Jung Il Lee
- Department of Orthopedic Surgery, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ji Hun Park
- Department of Orthopedic Surgery, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yeong-Rim Kim
- Medical Sensor Biomaterial Research Institute, Kwangwoon University, Seoul, Republic of Korea
| | - Kihak Gwon
- Medical Sensor Biomaterial Research Institute, Kwangwoon University, Seoul, Republic of Korea
| | - Hae Won Hwang
- Center for Biomaterials, Korea Institute of Science & Technology; Department of Materials Science and Engineering, Seoul National University, Seoul, Republic of Korea
| | - Gayoung Jung
- Center for Biomaterials, Korea Institute of Science & Technology, Seoul, Republic of Korea
| | - Joo-Yup Lee
- Department of Orthopedic Surgery, College of Medicine, Catholic University, Seoul, Republic of Korea
| | - Jeong-Yun Sun
- Department of Materials Science and Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jong Woong Park
- Department of Orthopedic Surgery, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Ho Shin
- Medical Sensor Biomaterial Research Institute; Department of Chemistry, Kwangwoon University, Seoul, Republic of Korea
| | - Myoung-Ryul Ok
- Center for Biomaterials, Korea Institute of Science & Technology, Seoul, Republic of Korea
| |
Collapse
|
4
|
Wang P, Ma K, Yang L, Zhang G, Ye M, Wang S, Wei S, Chen Z, Gu J, Zhang L, Niu J, Tao S. Predicting signaling pathways regulating demyelination in a rat model of lithium-pilocarpine-induced acute epilepsy: A proteomics study. Int J Biol Macromol 2021; 193:1457-1470. [PMID: 34742844 DOI: 10.1016/j.ijbiomac.2021.10.209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022]
Abstract
Demyelination is observed in animal models of intractable epilepsy (IE). Epileptogenesis damages the myelin sheath and dysregulates oligodendrocyte precursor cell (OPC) development. However, the molecular pathways regulating demyelination in epilepsy are unclear. Here, we predicted the molecular mechanisms regulating demyelination in a rat model of lithium-pilocarpine hydrochloride-induced epilepsy. We identified DGKA/Mboat2/Inpp5j and NOS/Keratin 28 as the main target molecules that regulate demyelination via glycerolipid and glycerophospholipid metabolism, phosphatidylinositol signaling, and estrogen signaling in demyelinated forebrain slice cultures (FSCs). In seizure-like FCSs, the actin cytoskeleton was regulated by Cnp and MBP via Pak4/Tmsb4x (also known as Tβ4) and Kif5c/Kntc1. Tβ4 possibly prevented OPC differentiation and maturation and inhibited MBP phosphorylation via the p38MAPK/ERK1/JNK1 pathway. The MAPK signaling pathway was more likely activated in seizure-like FCSs than in demyelinated FCSs. pMBP expression was decreased in the hippocampus of lithium-pilocarpine hydrochloride-induced acute epilepsy rats. The expression of remyelination-related factors was suppressed in the hippocampus and corpus callosum in lithium-pilocarpine hydrochloride-induced epilepsy rats. These findings suggest that the actin cytoskeleton, Tβ4, and MAPK signaling pathways regulate the decrease in pMBP in the hippocampus in a rat model of epilepsy. Our results indicate that regulating the actin cytoskeleton, Tβ4, and MAPK signaling pathways may facilitate the prevention of demyelination in IE.
Collapse
Affiliation(s)
- Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China.
| | - Kang Ma
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Lu Yang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Guodong Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Mengyi Ye
- Ningxia Medical University College of Traditional Chinese Medicine, Yinchuan 750004, Ningxia, China
| | - Siqi Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Shuangshuang Wei
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Zhangping Chen
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Jinghai Gu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Lianxiang Zhang
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China.
| | - Sun Tao
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China; Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
5
|
Panthi S, Gautam K. Roles of nitric oxide and ethyl pyruvate after peripheral nerve injury. Inflamm Regen 2017; 37:20. [PMID: 29259719 PMCID: PMC5725928 DOI: 10.1186/s41232-017-0051-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 09/01/2017] [Indexed: 12/27/2022] Open
Abstract
Short-lived reactive nitrogen species and reactive oxygen species have acquired significant attention in the field of biomedical science. Nitric oxide (NO), which was thought to be an unstable gas and pollutant, is now regarded as a gas transmitter like H2S and CO. NO is synthesized inside the mammalian body by l-arginine via three different isoforms of NO synthase whereas pyruvate is a glycolysis product and substrate for TCA cycle. Due to poor solubility and stability, therapeutic potential of pyruvate is limited. Ethyl pyruvate (EP) is now considered as a suitable replacement of pyruvate. In this paper, we will try to focus the effect of NO and EP in Schwann cell dedifferentiation, proliferation, nerve degeneration, and regeneration during Wallerian degeneration (WD) of peripheral nerve injury along with their neuroprotective effects, cardiovascular functioning, support in hepatic complication, etc.
Collapse
Affiliation(s)
- Sandesh Panthi
- Otago School of Biomedical Sciences, University of Otago, Otago, New Zealand
| | - Kripa Gautam
- China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
6
|
Gamage KK, Cheng I, Park RE, Karim MS, Edamura K, Hughes C, Spano AJ, Erisir A, Deppmann CD. Death Receptor 6 Promotes Wallerian Degeneration in Peripheral Axons. Curr Biol 2017; 27:890-896. [PMID: 28285993 DOI: 10.1016/j.cub.2017.01.062] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/19/2016] [Accepted: 01/30/2017] [Indexed: 11/17/2022]
Abstract
Axon degeneration during development is required to sculpt a functional nervous system and is also a hallmark of pathological insult, such as injury [1, 2]. Despite similar morphological characteristics, very little overlap in molecular mechanisms has been reported between pathological and developmental degeneration [3-5]. In the peripheral nervous system (PNS), developmental axon pruning relies on receptor-mediated extrinsic degeneration mechanisms to determine which axons are maintained or degenerated [5-7]. Receptors have not been implicated in Wallerian axon degeneration; instead, axon autonomous, intrinsic mechanisms are thought to be the primary driver for this type of axon disintegration [8-10]. Here we survey the role of neuronally expressed, paralogous tumor necrosis factor receptor super family (TNFRSF) members in Wallerian degeneration. We find that an orphan receptor, death receptor 6 (DR6), is required to drive axon degeneration after axotomy in sympathetic and sensory neurons cultured in microfluidic devices. We sought to validate these in vitro findings in vivo using a transected sciatic nerve model. Consistent with the in vitro findings, DR6-/- animals displayed preserved axons up to 4 weeks after injury. In contrast to phenotypes observed in Wlds and Sarm1-/- mice, preserved axons in DR6-/- animals display profound myelin remodeling. This indicates that deterioration of axons and myelin after axotomy are mechanistically distinct processes. Finally, we find that JNK signaling after injury requires DR6, suggesting a link between this novel extrinsic pathway and the axon autonomous, intrinsic pathways that have become established for Wallerian degeneration.
Collapse
Affiliation(s)
- Kanchana K Gamage
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Irene Cheng
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Rachel E Park
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Mardeen S Karim
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Kazusa Edamura
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Christopher Hughes
- Department of Physics and Astronomy, James Madison University, Harrisonburg, VA 22807, USA
| | - Anthony J Spano
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Alev Erisir
- Department of Psychology, University of Virginia, Charlottesville, VA 22903, USA
| | | |
Collapse
|
7
|
Maggio DM, Singh A, Iorgulescu JB, Bleicher DH, Ghosh M, Lopez MM, Tuesta LM, Flora G, Dietrich WD, Pearse DD. Identifying the Long-Term Role of Inducible Nitric Oxide Synthase after Contusive Spinal Cord Injury Using a Transgenic Mouse Model. Int J Mol Sci 2017; 18:ijms18020245. [PMID: 28125047 PMCID: PMC5343782 DOI: 10.3390/ijms18020245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/05/2017] [Accepted: 01/15/2017] [Indexed: 02/07/2023] Open
Abstract
Inducible nitric oxide synthase (iNOS) is a potent mediator of oxidative stress during neuroinflammation triggered by neurotrauma or neurodegeneration. We previously demonstrated that acute iNOS inhibition attenuated iNOS levels and promoted neuroprotection and functional recovery after spinal cord injury (SCI). The present study investigated the effects of chronic iNOS ablation after SCI using inos-null mice. iNOS-/- knockout and wild-type (WT) control mice underwent a moderate thoracic (T8) contusive SCI. Locomotor function was assessed weekly, using the Basso Mouse Scale (BMS), and at the endpoint (six weeks), by footprint analysis. At the endpoint, the volume of preserved white and gray matter, as well as the number of dorsal column axons and perilesional blood vessels rostral to the injury, were quantified. At weeks two and three after SCI, iNOS-/- mice exhibited a significant locomotor improvement compared to WT controls, although a sustained improvement was not observed during later weeks. At the endpoint, iNOS-/- mice showed significantly less preserved white and gray matter, as well as fewer dorsal column axons and perilesional blood vessels, compared to WT controls. While short-term antagonism of iNOS provides histological and functional benefits, its long-term ablation after SCI may be deleterious, blocking protective or reparative processes important for angiogenesis and tissue preservation.
Collapse
Affiliation(s)
- Dominic M Maggio
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institute of Heath, Bethesda, MD 20824, USA.
| | - Amanpreet Singh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - J Bryan Iorgulescu
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Drew H Bleicher
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Michael M Lopez
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Luis M Tuesta
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | - Govinder Flora
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
8
|
Nitric Oxide: Exploring the Contextual Link with Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7205747. [PMID: 28096943 PMCID: PMC5209623 DOI: 10.1155/2016/7205747] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/01/2016] [Indexed: 02/07/2023]
Abstract
Neuronal inflammation is a systematically organized physiological step often triggered to counteract an invading pathogen or to rid the body of damaged and/or dead cellular debris. At the crux of this inflammatory response is the deployment of nonneuronal cells: microglia, astrocytes, and blood-derived macrophages. Glial cells secrete a host of bioactive molecules, which include proinflammatory factors and nitric oxide (NO). From immunomodulation to neuromodulation, NO is a renowned modulator of vast physiological systems. It essentially mediates these physiological effects by interacting with cyclic GMP (cGMP) leading to the regulation of intracellular calcium ions. NO regulates the release of proinflammatory molecules, interacts with ROS leading to the formation of reactive nitrogen species (RNS), and targets vital organelles such as mitochondria, ultimately causing cellular death, a hallmark of many neurodegenerative diseases. AD is an enervating neurodegenerative disorder with an obscure etiology. Because of accumulating experimental data continually highlighting the role of NO in neuroinflammation and AD progression, we explore the most recent data to highlight in detail newly investigated molecular mechanisms in which NO becomes relevant in neuronal inflammation and oxidative stress-associated neurodegeneration in the CNS as well as lay down up-to-date knowledge regarding therapeutic approaches targeting NO.
Collapse
|
9
|
Rabinovich D, Yaniv SP, Alyagor I, Schuldiner O. Nitric Oxide as a Switching Mechanism between Axon Degeneration and Regrowth during Developmental Remodeling. Cell 2016; 164:170-182. [PMID: 26771490 DOI: 10.1016/j.cell.2015.11.047] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/02/2015] [Accepted: 11/13/2015] [Indexed: 12/31/2022]
Abstract
During development, neurons switch among growth states, such as initial axon outgrowth, axon pruning, and regrowth. By studying the stereotypic remodeling of the Drosophila mushroom body (MB), we found that the heme-binding nuclear receptor E75 is dispensable for initial axon outgrowth of MB γ neurons but is required for their developmental regrowth. Genetic experiments and pharmacological manipulations on ex-vivo-cultured brains indicate that neuronally generated nitric oxide (NO) promotes pruning but inhibits regrowth. We found that high NO levels inhibit the physical interaction between the E75 and UNF nuclear receptors, likely accounting for its repression of regrowth. Additionally, NO synthase (NOS) activity is downregulated at the onset of regrowth, at least partially, by short inhibitory NOS isoforms encoded within the NOS locus, indicating how NO production could be developmentally regulated. Taken together, these results suggest that NO signaling provides a switching mechanism between the degenerative and regenerative states of neuronal remodeling.
Collapse
Affiliation(s)
- Dana Rabinovich
- Department of Molecular Cell Biology, Weizmann Institute of Sciences, Rehovot 7610001, Israel
| | - Shiri P Yaniv
- Department of Molecular Cell Biology, Weizmann Institute of Sciences, Rehovot 7610001, Israel
| | - Idan Alyagor
- Department of Molecular Cell Biology, Weizmann Institute of Sciences, Rehovot 7610001, Israel
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Weizmann Institute of Sciences, Rehovot 7610001, Israel.
| |
Collapse
|
10
|
Palus K, Całka J. Alterations of neurochemical expression of the coeliac-superior mesenteric ganglion complex (CSMG) neurons supplying the prepyloric region of the porcine stomach following partial stomach resection. J Chem Neuroanat 2015; 72:25-33. [PMID: 26730724 DOI: 10.1016/j.jchemneu.2015.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 12/24/2022]
Abstract
The purpose of the present study was to determine the response of the porcine coeliac-superior mesenteric ganglion complex (CSMG) neurons projecting to the prepyloric area of the porcine stomach to peripheral neuronal damage following partial stomach resection. To identify the sympathetic neurons innervating the studied area of stomach, the neuronal retrograde tracer Fast Blue (FB) was applied to control and partial stomach resection (RES) groups. On the 22nd day after FB injection, following laparotomy, the partial resection of the previously FB-injected stomach prepyloric area was performed in animals of RES group. On the 28th day, all animals were re-anaesthetized and euthanized. The CSMG complex was then collected and processed for double-labeling immunofluorescence. In control animals, retrograde-labelled perikarya were immunoreactive to tyrosine hydroxylase (TH), dopamine β-hydroxylase (DβH), neuropeptide Y (NPY) and galanin (GAL). Partial stomach resection decreased the numbers of FB-positive neurons immunopositive for TH and DβH. However, the strong increase of NPY and GAL expression, as well as de novo-synthesis of neuronal nitric oxide synthase (nNOS) and leu5-Enkephalin (LENK) was noted in studied neurons. Furthermore, FB-positive neurons in all pigs were surrounded by a network of cocaine- and amphetamine-regulated transcript peptide (CART)-, calcitonin gene-related peptide (CGRP)-, and substance P (SP)-, vasoactive intestinal peptide (VIP)-, LENK- and nNOS- immunoreactive nerve fibers. This may suggest neuroprotective contribution of these neurotransmitters in traumatic responses of sympathetic neurons to peripheral axonal damage.
Collapse
Affiliation(s)
- Katarzyna Palus
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str. 13, 10- 718 Olsztyn, Poland.
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str. 13, 10- 718 Olsztyn, Poland.
| |
Collapse
|
11
|
Hippocampal Pruning as a New Theory of Schizophrenia Etiopathogenesis. Mol Neurobiol 2015; 53:2065-2081. [PMID: 25902861 DOI: 10.1007/s12035-015-9174-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/13/2015] [Indexed: 12/20/2022]
Abstract
Pruning in neurons has been suggested to be strongly involved in Schizophrenia's (SKZ) etiopathogenesis in recent biological, imaging, and genetic studies. We investigated the impact of protein-coding genes known to be involved in pruning, collected by a systematic literature research, in shaping the risk for SKZ in a case-control sample of 9,490 subjects (Psychiatric Genomics Consortium). Moreover, their modifications through evolution (humans, chimpanzees, and rats) and subcellular localization (as indicative of their biological function) were also investigated. We also performed a biological pathways (Gene Ontology) analysis. Genetics analyses found four genes (DLG1, NOS1, THBS4, and FADS1) and 17 pathways strongly involved in pruning and SKZ in previous literature findings to be significantly associated with the sample under analysis. The analysis of the subcellular localization found that secreted genes, and so regulatory ones, are the least conserved through evolution and also the most associated with SKZ. Their cell line and regional brain expression analysis found that their areas of primary expression are neuropil and the hippocampus, respectively. At the best of our knowledge, for the first time, we were able to describe the SKZ neurodevelopmental hypothesis starting from a single biological process. We can also hypothesize how alterations in pruning fine regulation and orchestration, strongly related with the evolutionary newest (and so more sensitive) secreted proteins, may be of particular relevance in the hippocampus. This early alteration may lead to a mis-structuration of neural connectivity, resulting in the different brain alteration that characterizes SKZ patients.
Collapse
|
12
|
S-Nitrosylation in neurogenesis and neuronal development. Biochim Biophys Acta Gen Subj 2014; 1850:1588-93. [PMID: 25527866 DOI: 10.1016/j.bbagen.2014.12.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/03/2014] [Accepted: 12/10/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Nitric oxide (NO) is a pleiotropic messenger molecule. The multidimensional actions of NO species are, in part, mediated by their redox nature. Oxidative posttranslational modification of cysteine residues to regulate protein function, termed S-nitrosylation, constitutes a major form of redox-based signaling by NO. SCOPE OF REVIEW S-Nitrosylation directly modifies a number of cytoplasmic and nuclear proteins in neurons. S-Nitrosylation modulates neuronal development by reaction with specific proteins, including the transcription factor MEF2. This review focuses on the impact of S-nitrosylation on neurogenesis and neuronal development. MAJOR CONCLUSIONS Functional characterization of S-nitrosylated proteins that regulate neuronal development represents a rapidly emerging field. Recent studies reveal that S-nitrosylation-mediated redox signaling plays an important role in several biological processes essential for neuronal differentiation and maturation. GENERAL SIGNIFICANCE Investigation of S-nitrosylation in the nervous system has elucidated new molecular and cellular mechanisms for neuronal development. S-Nitrosylated proteins in signaling networks modulate key events in brain development. Dysregulation of this redox-signaling pathway may contribute to neurodevelopmental disabilities such as autism spectrum disorder (ASD). Thus, further elucidation of the involvement of S-nitrosylation in brain development may offer potential therapeutic avenues for neurodevelopmental disorders. This article is part of a Special Issue entitled Redox regulation of differentiation and de-differentiation.
Collapse
|
13
|
Effects of cerebrolysin on rat Schwann cells in vitro. Acta Histochem 2014; 116:820-30. [PMID: 24636538 DOI: 10.1016/j.acthis.2014.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 12/11/2022]
Abstract
Although the peripheral nervous system (PNS) is capable of regeneration, these processes are limited. As a potential means to augment PNS regeneration, the effects of cerebrolysin (CL), a proteolytic peptide fraction, were tested in vitro on Schwann cell (SC) proliferation, stress resistance, phagocytic and cluster-forming capacity. Primary SC/fibrocyte co-cultures were prepared from dorsal root ganglia of 5-7-day-old rats. SCs were subjected to mechanical stress by media change and metabolic stress by serum glucose deprivation (SGD). Cell survival was assessed using MTT test. SC proliferation was determined by counting BrdU-labeled cells. SC clustering was studied by ImageJ analysis of S100 immunostaining. Wallerian degeneration (WD) was evaluated by measuring acetylcholine-esterase staining within sciatic nerves in vitro. It was found that CL caused no effect on MTT turnover in the tested doses. CL inhibited SC proliferation in a dose-dependent manner. Media change and additional SGD stress inhibited SC clustering. CL enhanced the reorganization of SC clusters and was able to counteract SGD-induced cluster defects. Moreover, CL accelerated WD in vitro. CL was able to enhance the functions of SCs that are relevant to nerve regeneration. Thus, our findings suggest that CL may be suitable for therapeutic usage to enhance PNS regeneration/reconstruction.
Collapse
|
14
|
Keilhoff G, Schröder H, Peters B, Becker A. Time-course of neuropathic pain in mice deficient in neuronal or inducible nitric oxide synthase. Neurosci Res 2013; 77:215-21. [DOI: 10.1016/j.neures.2013.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/22/2013] [Accepted: 08/24/2013] [Indexed: 10/26/2022]
|
15
|
Nitric oxide synthesis and cGMP production is important for neurite growth and synapse remodeling after axotomy. J Neurosci 2013; 33:5626-37. [PMID: 23536077 DOI: 10.1523/jneurosci.3659-12.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nitric oxide (NO) is an important signaling molecule with a variety of functions in the CNS, including a potential role in modulating neuronal growth and synapse formation. In the present study, we used tractable, identified neurons in the CNS of the pond snail Lymnaea stagnalis to study the role of endogenous NO signaling in neuronal growth and synaptic remodeling after nerve injury. Axonal damage of L. stagnalis neurons B1 and B2 induces extensive central growth of neurites that is accompanied by changes in existing electrical connections, the transient formation of novel electrical connections, and the formation of a novel excitatory chemical synapse from B2 to B1 neurons. Partial chronic inhibition of endogenous NO synthesis reduces neurite growth in NO-synthase-expressing B2, but has only minor effects on NOS-negative B1 neurons. Chronic application of an NO donor while inhibiting endogenous NO synthesis rescues neurite extension in B2 neurons and boosts growth of B1 neurons. Blocking soluble guanylate cyclase activity completely suppresses neurite extension and synaptic remodeling after nerve crush, demonstrating the importance of cGMP in these processes. Interestingly, inhibition of cGMP-dependent protein kinase only suppresses chemical synapse formation without effects on neuronal growth and electrical synapse remodeling. We conclude that NO signaling via cGMP is an important modulator of both neurite growth and synaptic remodeling after nerve crush. However, differential effects of cGMP-dependent protein kinase inhibition on neurite growth and synaptic remodeling suggest that these effects are mediated by separate signaling pathways.
Collapse
|
16
|
Sezen SF, Lagoda G, Burnett AL. Neuronal nitric oxide signaling regulates erection recovery after cavernous nerve injury. J Urol 2011; 187:757-63. [PMID: 22177198 DOI: 10.1016/j.juro.2011.09.146] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Indexed: 12/31/2022]
Abstract
PURPOSE Nitric oxide is the major neuronal mediator of penile erection but its role in erectile function status after cavernous nerve injury is uncertain. We determined the function of neuronal nitric oxide signaling in the pathobiology of erectile function recovery after partial cavernous nerve injury using genetic and pharmacological mouse experimental paradigms. MATERIALS AND METHODS Erectile function was evaluated in 5 to 7 wild-type and neuronal nitric oxide synthase-α knockout mice per group 1, 3 and 7 days after unilateral crush or sham injury, at day 7 in wild-type mice treated with the nitric oxide synthase inhibitor L-NAME (l-nitro arginine methyl ester) (Sigma-Aldrich®) at baseline and for 6 days after unilateral crush injury. Apoptosis in the penis was evaluated by Western blot analysis of p-Akt-S473, 3-nitrotyrosine and caspase-3 after bilateral crush injury. RESULTS Intracavernous pressure was significantly decreased at 1, 3 and 7 days in wild-type mice but only at day 1 in knockout mice after unilateral crush injury compared with sham treatment values (p <0.05). L-NAME treated wild-type mice had improved erectile function compared with the vehicle treated group at day 7 after unilateral crush injury (p <0.05). In penes p-Akt-S473 was significantly decreased in vehicle treated (p <0.05) but not in L-NAME treated wild-type mice. In penes 3-nitrotyrosine was significantly decreased in L-NAME treated wild-type and vehicle treated knockout mice (p <0.05). Caspase-3 in penes was significantly increased in vehicle treated (p <0.05) but not in L-NAME treated wild-type mice and vehicle treated knockout mice. CONCLUSIONS Neuronal nitric oxide signaling regulates erectile function recovery early after partial cavernous nerve injury, exerting an inhibitory role via the induction of apoptotic change in penile tissue. Therapeutic strategies to improve erectile function recovery after radical prostatectomy may consider targeting pathogenic sites of nitric oxide neurobiology.
Collapse
Affiliation(s)
- Sena F Sezen
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | |
Collapse
|
17
|
Abstract
Traditionally, researchers have believed that axons are highly dependent on their cell bodies for long-term survival. However, recent studies point to the existence of axon-autonomous mechanism(s) that regulate rapid axon degeneration after axotomy. Here, we review the cellular and molecular events that underlie this process, termed Wallerian degeneration. We describe the biphasic nature of axon degeneration after axotomy and our current understanding of how Wld(S)--an extraordinary protein formed by fusing a Ube4b sequence to Nmnat1--acts to protect severed axons. Interestingly, the neuroprotective effects of Wld(S) span all species tested, which suggests that there is an ancient, Wld(S)-sensitive axon destruction program. Recent studies with Wld(S) also reveal that Wallerian degeneration is genetically related to several dying back axonopathies, thus arguing that Wallerian degeneration can serve as a useful model to understand, and potentially treat, axon degeneration in diverse traumatic or disease contexts.
Collapse
Affiliation(s)
- Michael P Coleman
- Laboratory of Molecular Signaling, The Babraham Institute, Cambridge CB223AT, United Kingdom
| | | |
Collapse
|
18
|
Abstract
Axons depend critically on axonal transport both for supplying materials and for communicating with cell bodies. This chapter looks at each activity, asking what aspects are essential for axon survival. Axonal transport declines in neurodegenerative disorders, such as Alzheimer's disease, amyotrophic lateral sclerosis, and multiple sclerosis, and in normal ageing, but whether all cargoes are equally affected and what limits axon survival remains unclear. Cargoes can be differentially blocked in some disorders, either individually or in groups. Each missing protein cargo results in localized loss-of-function that can be partially modeled by disrupting the corresponding gene, sometimes with surprising results. The axonal response to losing specific proteins also depends on the rates of protein turnover and on whether the protein can be locally synthesized. Among cargoes with important axonal roles are components of the PI3 kinase, Mek/Erk, and Jnk signaling pathways, which help to communicate with cell bodies and to regulate axonal transport itself. Bidirectional trafficking of Bdnf, NT-3, and other neurotrophic factors contribute to intra- and intercellular signaling, affecting the axon's cellular environment and survival. Finally, several adhesion molecules and gangliosides are key determinants of axon survival, probably by mediating axon-glia interactions. Thus, failure of long-distance intracellular transport can deprive axons of one, few, or many cargoes. This can lead to axon degeneration either directly, through the absence of essential axonal proteins, or indirectly, through failures in communication with cell bodies and nonneuronal cells.
Collapse
|
19
|
Martínez de Albornoz P, Delgado PJ, Forriol F, Maffulli N. Non-surgical therapies for peripheral nerve injury. Br Med Bull 2011; 100:73-100. [PMID: 21429947 DOI: 10.1093/bmb/ldr005] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Non-surgical approaches have been developed to enhance nerve recovery, which are complementary to surgery and are an adjunct to the reinnervation process. SOURCES OF DATA A search of PubMed, Medline, CINAHL, DH data and Embase databases was performed using the keywords 'peripheral nerve injury' and 'treatment'. AREAS OF CONTROVERSY Most of the conservative therapies are focused to control neuropathic pain after nerve tissue damage. Only physical therapy modalities have been studied in humans and their effectiveness is not proved. GROWING POINTS Many modalities have been experimented with to promote nerve healing and restore function in animal models and in vitro studies. Despite this, none have been actually translated into clinical practice. AREAS TIMELY FOR DEVELOPING RESEARCH The hypotheses proved in animals and in vitro should be translated to human clinical practice.
Collapse
Affiliation(s)
- Pilar Martínez de Albornoz
- Department of Trauma and Orthopaedic Surgery, FREMAP Hospital, Ctra de Pozuelo 61, 28220 Majadahonda, Madrid, Spain
| | | | | | | |
Collapse
|
20
|
Moreno-López B. Local isoform-specific NOS inhibition: a promising approach to promote motor function recovery after nerve injury. J Neurosci Res 2010; 88:1846-57. [PMID: 20143424 DOI: 10.1002/jnr.22353] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Physical injury to a nerve is the most frequent cause of acquired peripheral neuropathy, which is responsible for loss of motor, sensory and/or autonomic functions. Injured axons in the peripheral nervous system maintain the capacity to regenerate in adult mammals. However, after nerve transection, stumps of damaged nerves must be surgically joined to guide regenerating axons into the distal nerve stump. Even so, severe functional limitations persist after restorative surgery. Therefore, the identification of molecules that regulate degenerative and regenerative processes is indispensable in developing therapeutic tools to accelerate and improve functional recovery. Here, I consider the role of nitric oxide (NO) synthesized by the three major isoforms of NO synthases (NOS) in motor neuropathy. Neuronal NOS (nNOS) seems to be the primary source of NO that is detrimental to the survival of injured motoneurons. Endothelial NOS (eNOS) appears to be the major source of NO that interferes with axonal regrowth, at least soon after injury. Finally, NO derived from inducible NOS (iNOS) or nNOS is critical to the process of lipid breakdown for Wallerian degeneration and thereby benefits axonal regrowth. Specific inhibitors of these isoforms can be used to protect injured neurons from degeneration and promote axonal regeneration. A cautious proposal for the treatment of acquired motor neuropathy using therapeutic tools that locally interfere with eNOS/nNOS activities seems to merit consideration.
Collapse
|
21
|
Stern M, Bicker G. Nitric oxide as a regulator of neuronal motility and regeneration in the locust embryo. JOURNAL OF INSECT PHYSIOLOGY 2010; 56:958-965. [PMID: 20361970 DOI: 10.1016/j.jinsphys.2010.03.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 03/18/2010] [Accepted: 03/19/2010] [Indexed: 05/29/2023]
Abstract
Nitric oxide (NO) is known as a gaseous messenger in the nervous system. It plays a role in synaptic plasticity, but also in development and regeneration of nervous systems. We have studied the function of NO and its signaling cascade via cyclic GMP in the locust embryo. Its developing nervous system is well suited for pharmacological manipulations in tissue culture. The components of this signaling pathway are localized by histochemical and immunofluorescence techniques. We have analyzed cellular mechanisms of NO action in three examples: 1. in the peripheral nervous system during antennal pioneer axon outgrowth, 2. in the enteric nervous system during migration of neurons forming the midgut nerve plexus, and 3. in the central nervous system during axonal regeneration of serotonergic neurons after axotomy. In each case, internally released NO or NO-induced cGMP synthesis act as permissive signals for the developmental process. Carbon monoxide (CO), as a second gaseous messenger, modulates enteric neuron migration antagonistic to NO.
Collapse
Affiliation(s)
- Michael Stern
- Division of Cell Biology, Institute of Physiology, University of Veterinary Medicine Hannover, D-30173 Hannover, Germany.
| | | |
Collapse
|
22
|
Sakakima H, Yoshida Y, Yamazaki Y, Matsuda F, Ikutomo M, Ijiri K, Muramatsu H, Muramatsu T, Kadomatsu K. Disruption of the midkine gene (Mdk) delays degeneration and regeneration in injured peripheral nerve. J Neurosci Res 2009; 87:2908-15. [DOI: 10.1002/jnr.22127] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
23
|
Sezen SF, Lagoda G, Burnett AL. Role of immunophilins in recovery of erectile function after cavernous nerve injury. J Sex Med 2009; 6 Suppl 3:340-6. [PMID: 19267858 DOI: 10.1111/j.1743-6109.2008.01193.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Immunophilin ligands provide potentially new alternatives for the treatment of erectile dysfunction (ED), which occurs after injury of the cavernous nerves (CNs). AIM To review and update current knowledge of the neurotrophic effects and likely mechanism of action of immunophilin proteins with emphasis on the FK506-binding protein (FKBP) subfamily and the role of immunophilin ligands for the treatment of CN injury-induced ED. METHODS Review of available reports of studies investigating the effects and neurotrophic mechanisms of immunophilin ligands involved in erectile function recovery in rodent models of CN injury. MAIN OUTCOME MEASURES Erection parameters and molecular correlations associated with CN injury and functional recovery. RESULTS Treatment with prototype immunosuppressive immunophilin ligands FK506 (FK) and rapamycin (Rapa) improve erectile function in animal models of CN injury. Similarly, non-immunosuppressive analogs such as GPI-1046 and FK1706 are effective in recovery of erections after CN injury. Neuronal nitric oxide may influence the erection recovery effects of immunophilin ligands after CN injury. FKBPs 38 and 65 expression changes in the penis and its innervation coincide with the neurotrophic effects of immunophilin ligands. Antioxidative actions of immunophilin ligands contribute to their neurotrophic effects. Immunophilins are localized to nerves coursing in human prostate and penile tissue. CONCLUSIONS The findings support the hypothesis that immunophilin ligands, working through specific receptor mechanisms that are specific to injured CN, are potentially useful to sustain erectile function in men following radical prostatectomy.
Collapse
Affiliation(s)
- Sena F Sezen
- James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Department of Urology, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
24
|
The Potential Role of Nitric Oxide Synthase in Survival and Regeneration of Magnocellular Neurons of Hypothalamo-Neurohypophyseal System. Neurochem Res 2009; 34:1907-13. [DOI: 10.1007/s11064-009-9965-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Accepted: 03/25/2009] [Indexed: 12/21/2022]
|
25
|
Inhibition of nitric oxide synthase promotes facial axonal regeneration following neurorrhaphy. Exp Neurol 2009; 216:499-510. [PMID: 19320008 DOI: 10.1016/j.expneurol.2009.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Reconnection of interrupted peripheral nerve by microsurgical suture is a common clinical practice. However, the extent to which peripheral neurorrhaphy improves nerve regeneration and functional recovery remains unsatisfactory. Here, we used anatomical and electrophysiological techniques to investigate the temporal correlation between the expressions of oxidative stress-related biomarkers such as neuronal nitric oxide synthase (nNOS) and the facial axonal regeneration after an immediate facial nerve repair in adult rats since peripheral nerve lesion is well known to induce a dramatic increase of NOS expression in the affected neuronal cell bodies. We found that compared to nerve cut without suture, facial nerve repair not only caused the facial axonal regeneration but also consistently prevented the fluctuations of expressions of oxidative stress-related biomarkers in 10 weeks postlesion. To further elucidate the role of nitric oxide (NO) in the axonal degeneration/regeneration, four different NOS inhibitors were applied to additional rats after facial nerve cut or repair. Both of facial nerve cut+NOS inhibition and facial nerve repair+NOS inhibition were seen to prevent the alterations of expressions of the biomarkers, no matter which NOS inhibitor was used. Moreover, we found that facial nerve repair+NOS inhibition promoted earlier and better axonal regeneration than facial nerve repair, demonstrated by labeling of neuromuscular junctions, retrograde tracing, and electromyography. These results provide direct evidence that peripheral nerve suture and/or treatment of NOS inhibitors can maintain the homeostasis of oxidative stress-related biomarkers, especially nNOS in neuronal cell bodies. These actions may thus facilitate the axonal regeneration.
Collapse
|
26
|
Zhou LH, Han S, Xie YY, Wang LL, Yao ZB. Differences in c-jun and nNOS expression levels in motoneurons following different kinds of axonal injury in adult rats. ACTA ACUST UNITED AC 2009; 36:213-27. [PMID: 19238548 DOI: 10.1007/s11068-009-9040-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 12/13/2008] [Accepted: 12/18/2008] [Indexed: 11/28/2022]
Abstract
In the peripheral nervous system (PNS), root avulsion causes motoneuron degeneration, but the majority of motoneurons can survive axotomy. In order to study the mechanism of motoneuron degeneration, we compared the expression patterns of c-jun and neuronal nitric oxide synthase (nNOS), the well-known molecular players in PNS regeneration and degeneration, among adult rats having undergone axotomy (Ax), avulsion (Av), or pre-axotomy plus secondary avulsion (Ax + Av) of the brachial plexus. Our results showed that the highest and longest-lasting c-jun activation occurred in Ax, which was much stronger than those in Av and Ax + Av. The time course and intensity of c-jun expression in Ax + Av were similar to those in Av except on day 1, while the pre-axotomy condition resulted in a transient up-regulation of c-jun to a level comparable to that in Ax. Axotomy alone did not induce nNOS expression in motoneurons. Pre-axotomy left-shifted the time course of nNOS induction in Ax + Av compared to that in Av. Motoneuron loss was not evident in Ax, while it was 70% in Av and more than 85% in Ax + Av at 8 weeks postinjury. The survival of motoneurons was positively correlated with c-jun induction, but not with nNOS expression in motoneurons. Moreover, c-jun induction was negatively correlated with nNOS induction in injured motoneurons. Our results indicate that functional crosstalk between c-jun and nNOS might play an important role in avulsion-induced motoneuron degeneration, while c-jun might act as a prerequisite survival factor and nNOS might act as a predictor for the onset of motoneuron degeneration.
Collapse
Affiliation(s)
- Li-Hua Zhou
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-sen University, Guangzhou 510080, PR China
| | | | | | | | | |
Collapse
|
27
|
Sunico CR, Portillo F, González-Forero D, Kasparov S, Moreno-López B. Evidence for a detrimental role of nitric oxide synthesized by endothelial nitric oxide synthase after peripheral nerve injury. Neuroscience 2008; 157:40-51. [PMID: 18824216 DOI: 10.1016/j.neuroscience.2008.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 09/02/2008] [Accepted: 09/03/2008] [Indexed: 12/17/2022]
Abstract
Physical injury to a nerve is the most common cause of acquired peripheral neuropathy. Identification of molecules involved in degenerative and regenerative processes is a key step toward development of therapeutic tools in order to accelerate motor, sensory and/or autonomic function recovery. We have studied the role of nitric oxide (NO) using as a model the severe crushing of a motor nerve in adult rats. This type of injury up-regulates the three isoforms of nitric oxide synthase (NOS) in the affected nerve. Chronic systemic inhibition of NOS accelerated the onset of functional muscle reinnervation evaluated by the recording of compound muscle action potential evoked by electrical stimulation of the injured nerve. Besides, it increased the number of back-labeled motoneurons by application, 2 days after injury, of a retrograde marker 10 mm distal to the crushing site. These effects were mimicked by chronic specific inhibition of the endothelial isoform of nitric oxide synthase (eNOS), but not by specific inhibitors of the neuronal or inducible isoform. Next, we intraneurally injected a replication-deficient adenoviral vector directing the expression of a dominant negative mutant of eNOS (Ad-TeNOS). A single injection of Ad-TeNOS on the day of crushing significantly accelerated functional recovery of neuromuscular junction and increased axonal regeneration. Moreover, Ad-TeNOS did not compromise motoneuron viability or stability of reestablished neuromuscular junctions. Taken together, these results suggest that NO of endothelial origin slows down muscle reinnervation by means of detrimental actions on axonal regeneration after peripheral nerve injury. These experiments identify eNOS as a potential therapeutic target for treatment of traumatic nerve injuries and highlight the potential of gene therapy in treating injuries of this type using viral vectors to suppress the activity of eNOS.
Collapse
Affiliation(s)
- C R Sunico
- Area de Fisiología, Facultad de Medicina, Universidad de Cádiz, Plaza Falla, 9, 11003 Cádiz, Spain
| | | | | | | | | |
Collapse
|
28
|
Spatiotemporal Expression of PSD-95 and nNOS After Rat Sciatic Nerve Injury. Neurochem Res 2007; 33:1090-100. [DOI: 10.1007/s11064-007-9555-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2007] [Accepted: 11/20/2007] [Indexed: 12/01/2022]
|
29
|
Stroissnigg H, Trancíková A, Descovich L, Fuhrmann J, Kutschera W, Kostan J, Meixner A, Nothias F, Propst F. S-nitrosylation of microtubule-associated protein 1B mediates nitric-oxide-induced axon retraction. Nat Cell Biol 2007; 9:1035-45. [PMID: 17704770 DOI: 10.1038/ncb1625] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 07/26/2007] [Indexed: 02/05/2023]
Abstract
Treatment of cultured vertebrate neurons with nitric oxide leads to growth-cone collapse, axon retraction and the reconfiguration of axonal microtubules. We show that the light chain of microtubule-associated protein (MAP) 1B is a substrate for S-nitrosylation in vivo, in cultured cells and in vitro. S-nitrosylation occurs at Cys 2457 in the COOH terminus. Nitrosylation of MAP1B leads to enhanced interaction with microtubules and correlates with the inhibition of neuroblastoma cell differentiation. We further show, in dorsal root ganglion neurons, that MAP1B is necessary for neuronal nitric oxide synthase control of growth-cone size, growth-cone collapse and axon retraction. These results reveal an S-nitrosylation-dependent signal-transduction pathway that is involved in regulation of the axonal cytoskeleton and identify MAP1B as a major component of this pathway. We propose that MAP1B acts by inhibiting a microtubule- and dynein-based mechanism that normally prevents axon retraction.
Collapse
Affiliation(s)
- Heike Stroissnigg
- Max F. Perutz Laboratories, Department of Molecular Cell Biology, University of Vienna, Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gray M, Palispis W, Popovich PG, van Rooijen N, Gupta R. Macrophage depletion alters the blood-nerve barrier without affecting Schwann cell function after neural injury. J Neurosci Res 2007; 85:766-77. [PMID: 17266098 DOI: 10.1002/jnr.21166] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Previous work has shown that, during the early phases of chronic nerve compression (CNC) injury, axonal pathology is absent while Schwann cells undergo a dramatic process of cellular turnover with marked proliferation. It is known that macrophages may release Schwann cell mitogens, so we sought to explore the role of macrophages in CNC injury by selectively depleting the population of hematogenously derived macrophages in nerves undergoing CNC injury by injecting clodronate liposomes at days 1, 3, and 6 postinjury and evaluating both the integrity of the blood-nerve barrier (BNB) and Schwann cell function. Integrity of the BNB was evaluated by intravenously injecting Evans blue albumin (EBA), and Schwann cell number was determined via stereologic techniques. The BNB was clearly altered by 2 weeks postinjury and continued to disintegrate at later time points. Macrophage depletion attenuated this response at all observed time points. Quantification of Schwann cell nuclei in CNC nerves showed no differences between compressed sections of macrophage-depleted and nondepleted animals. Although macrophages are largely responsible for the increased vascular permeability associated with CNC injury, it is likely that the Schwann cell response to CNC injury is not influenced by macrophage-derived mitogenic signals but rather must be mediated via alternative mechanisms.
Collapse
Affiliation(s)
- Michael Gray
- Department of Orthopaedic Surgery, University of California, Irvine, Irvine, California, USA
| | | | | | | | | |
Collapse
|
31
|
Bicker G. Pharmacological approaches to nitric oxide signalling during neural development of locusts and other model insects. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2007; 64:43-58. [PMID: 17167749 DOI: 10.1002/arch.20161] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A novel aspect of cellular signalling during the formation of the nervous system is the involvement of the messenger molecule nitric oxide (NO), which has been discovered in the mammalian vascular system as mediator of smooth muscle relaxation. NO is a membrane-permeant molecule, which activates soluble guanylyl cyclase (sGC) and leads to the formation of cyclic GMP (cGMP) in target cells. The analysis of specific cell types in model insects such as Locusta, Schistocerca, Acheta, Manduca, and Drosophila shows that the NO/cGMP pathway is required for the stabilization of photoreceptor growth cones at the start of synaptic assembly in the optic lobe, for regulation of cell proliferation, and for correct outgrowth of pioneer neurons. Inhibition of the NOS and sGC enzymes combined with rescue experiments show that NO, and potentially also another atypical messenger, carbon monoxide (CO), orchestrate cell migration of enteric neurons. Cultured insect embryos are accessible model systems in which the molecular pathways linking cytoskeletal rearrangement to directed cell movements can be analyzed in natural settings. Based on the results obtained from the insect models, I discuss current evidence for NO and cGMP as essential signalling molecules for the development of vertebrate brains.
Collapse
Affiliation(s)
- Gerd Bicker
- University of Veterinary Medicine Hannover, Cell Biology, Institute of Physiology, Hannover, Germany.
| |
Collapse
|
32
|
Alluin O, Feron F, Desouches C, Dousset E, Pellissier JF, Magalon G, Decherchi P. Metabosensitive Afferent Fiber Responses after Peripheral Nerve Injury and Transplantation of an Acellular Muscle Graft in Association with Schwann Cells. J Neurotrauma 2006; 23:1883-94. [PMID: 17184196 DOI: 10.1089/neu.2006.23.1883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Studies dedicated to the repair of peripheral nerve focused almost exclusively on motor or mechanosensitive fiber regeneration. Poor attention has been paid to the metabosensitive fibers from group III and IV (also called ergoreceptor). Previously, we demonstrated that the metabosensitive response from the tibialis anterior muscle was partially restored when the transected nerve was immediately sutured. In the present study, we assessed motor and metabosensitive responses of the regenerated axons in a rat model in which 1 cm segment of the peroneal nerve was removed and immediately replaced by an autologous nerve graft or an acellular muscle graft. Four groups of animals were included: control animals (C, no graft), transected animals grafted with either an autologous nerve graft (Gold Standard-GS) or an acellular muscle filled with Schwann Cells (MSC) or Culture Medium (MCM). We observed that (1) the tibialis anterior muscle was atrophied in GS, M(SC) and M(CM) groups, with no significant difference between grafted groups; (2) the contractile properties of the reinnervated muscles after nerve stimulation were similar in all groups; (3) the metabosensitive afferent responses to electrically induced fatigue was smaller in M(SC) and MCM groups; and (4) the metabosensitive afferent responses to two chemical agents (KCl and lactic acid) was decreased in GS, M(SC) and M(CM) groups. Altogether, these data indicate a motor axonal regeneration and an immature metabosensitive afferent fiber regrowth through acellular muscle grafts. Similarities between the two groups grafted with acellular muscles suggest that, in our conditions, implanted Schwann cells do not improve nerve regeneration. Future studies could include engineered conduits that mimic as closely as possible the internal organization of uninjured nerve.
Collapse
Affiliation(s)
- Olivier Alluin
- Laboratoire des Déterminants Physiologiques de l'Activité Physique (UPRES EA 3285), Institut Fédératif de Recherche (IFR) 107, Faculté des Sciences du Sport, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Many illnesses that affect the peripheral nervous system (PNS) lead to distal axonal degeneration rather than loss of neuronal cell bodies. Strategies aimed at promoting survival of injured neurons (i.e., preventing cell death) may not be applicable to many PNS illnesses. We have developed in vitro and in vivo animal models to study mechanisms of acquired peripheral neuropathies and used these models to evaluate the therapeutic potential of novel compounds. In recent years, erythropoietin (EPO) has been recognized as a novel neuroprotectant in the central nervous system. In the PNS, we recently showed that Schwann cell-derived EPO acts as an endogenous neuroprotectant and that it is most effective in preventing distal axonal degeneration seen in models of peripheral neuropathy. Similarly, we showed that immunophilin ligands are also neuroprotective in the PNS and prevent axonal degeneration seen in models of peripheral neuropathies. Both EPO and non-immunosuppressive immunophilin ligands are in early clinical development for the treatment of acquired peripheral neuropathies.
Collapse
Affiliation(s)
- Ahmet Höke
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Path 509, Baltimore, Maryland 21287, USA.
| | | |
Collapse
|
34
|
Jungnickel J, Haase K, Konitzer J, Timmer M, Grothe C. Faster nerve regeneration after sciatic nerve injury in mice over-expressing basic fibroblast growth factor. ACTA ACUST UNITED AC 2006; 66:940-8. [PMID: 16758491 DOI: 10.1002/neu.20265] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Basic fibroblast growth factor (FGF-2) is expressed in the peripheral nervous system and is up-regulated after nerve lesion. It has been demonstrated that administration of FGF-2 protects neurons from injury-induced cell death and promotes axonal regrowth. Using transgenic mice over-expressing FGF-2 (TgFGF-2), we addressed the importance of endogenously generated FGF-2 on sensory neuron loss and sciatic nerve regeneration. After sciatic nerve transection, wild-type and transgenic mice showed the same degree of cell death in L5 spinal ganglia. Also, the number of chromatolytic, eccentric, and pyknotic sensory neurons was not changed under elevated levels of FGF-2. Morphometric evaluation of intact nerves from TgFGF-2 mice revealed no difference in number and size of myelinated fibers compared to wild-type mice. One week after crush injury, the number of regenerated axons was doubled and the myelin thickness was significantly smaller in transgenic mice. After 2 and 4 weeks, morphometric analysis and functional tests revealed no differences in recovery of sensory and motor nerve fibers. To study the role of FGF-2 over-expression on Schwann cell proliferation during the early regeneration process, we used BrdU-labeling to mark dividing cells. In transgenic mice, the number of proliferating cells was significantly increased distal to the crush site compared to wild-types. We propose that endogenously synthesized FGF-2 influences early peripheral nerve regeneration by regulating Schwann cell proliferation, axonal regrowth, and remyelination.
Collapse
Affiliation(s)
- Julia Jungnickel
- Department of Neuroanatomy OE 4140, Center of Anatomy, Hannover Medical School, D-30623 Hannover, Germany.
| | | | | | | | | |
Collapse
|
35
|
Zhou L, Wu W. Antisense oligos to neuronal nitric oxide synthase aggravate motoneuron death induced by spinal root avulsion in adult rat. Exp Neurol 2005; 197:84-92. [PMID: 16246329 DOI: 10.1016/j.expneurol.2005.08.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 08/02/2005] [Accepted: 08/11/2005] [Indexed: 10/25/2022]
Abstract
The present study used nitric oxide synthase (nNOS) antisense oligos (nNOS AS-ODN) to assess the role of nNOS in motoneuron death induced by spinal root avulsion. A right seventh cervical (C7) spinal root avulsion was performed on adult male Sprague-Dawley rats. Two weeks later, FITC-labeled random oligos (FITC-R-ODN), nNOS AS-ODN, R-ODN or TE buffer was applied to the lesioned side of the C7 spinal segment and refreshed every 3 days. FITC-R-ODN was first detected inside the injured motoneurons at 10 h, accumulated to a maximum by 24 h and faded out from 72 h. Following avulsion, nNOS AS-ODN decreased the number of nNOS-positive motoneurons in the lesioned segment compared either with buffer (P < 0.001 at 15 days, 3 and 4 weeks post-injury) or with R-ODN control (P = 0.002 at 15 days, P < 0.001 at 3 and 4 weeks post-injury). Interestingly, nNOS AS-ODN also decreased the number of surviving motoneurons compared either with buffer (P = 0.005 at 15 days, P < 0.001 at 3 or 4 weeks) or with R-ODN control (P < 0.001 at 3 or 4 weeks). Meanwhile, there were no significant differences between R-ODN and buffer control either in the number of nNOS-positive motoneurons (P = 0.245 at 15 days, P = 0.089 at 3 weeks and P = 0.162 at 4 weeks) or in the number of surviving motoneurons (P = 0.426 at 15 days, P = 0.321 at 3 weeks or P = 0.344 at 4 weeks). These findings indicate that nNOS AS-ODN, applied from 2 weeks after avulsion, aggravates the motoneuron death due to root avulsion by specifically down-regulating nNOS gene expression and that the expression of nNOS in adult spinal motoneurons in response to root avulsion may play a beneficial role in the survival of injured neurons.
Collapse
Affiliation(s)
- Lihua Zhou
- Department of Anatomy, Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | | |
Collapse
|
36
|
Cunningham MG, Donalds RA, Scouten CW, Tresch MC. A versatile, low-cost adaptor for stereotaxic and electrophysiologic spinal preparations in juvenile and adult rodents. Brain Res Bull 2005; 68:157-62. [PMID: 16325015 DOI: 10.1016/j.brainresbull.2005.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2005] [Revised: 07/19/2005] [Accepted: 08/08/2005] [Indexed: 11/23/2022]
Abstract
Rats and mice provide excellent models for normal spinal cord physiology, traumatic spinal cord injury, and various disease states. Alternative and improved methodologies for experimental spinal preparations are desirable, particularly in the wake of expanding neuroscience technology, such as the diverse array of transgenic mice now available, and exciting new therapeutic approaches, including transplantation and gene therapy. This report describes a simple, low-cost instrument for spinal preparations in rodents of different sizes, including rat pups. The device adapts to standard small animal stereotaxic instruments, precluding the need for additional stereotaxic apparatus. Surgical methods utilizing the device are presented demonstrating the instrument's capacity for precise alignment and stabilization of the spinal column that is reproducible from animal to animal. Proof of concept is demonstrated with results from spinal cord injections and electrophysiologic recordings.
Collapse
Affiliation(s)
- Miles G Cunningham
- Laboratory for Neural Reconstruction, McLean Hospital, Program in Neuroscience and Department of Psychiatry, Harvard Medical School, Boston, MA 02478, USA.
| | | | | | | |
Collapse
|
37
|
Abstract
Compressive neuropathies are highly prevalent, debilitating conditions with variable functional recovery after surgical decompression. Chronic nerve compression injury induces concurrent Schwann cell proliferation and apoptosis in the early stages of the disorder, independent of axonal injury. These proliferating Schwann cells locally demyelinate and remyelinate in the region of injury. Furthermore, Schwann cells upregulate vascular endothelial growth factor secondary to chronic nerve compression injury and induce neovascularization to facilitate the recruitment of macrophages. In contrast to Wallerian degeneration, macrophage recruitment occurs gradually with chronic nerve compression injury and continues for a longer time. Schwann cells change their gene and protein expression in response to mechanical stimuli as shear stress decreases the expression of myelin associated glycoprotein and myelin basic protein mRNA and protein for in vitro promyelinating Schwann cells. The local down-regulation of myelin associated glycoprotein in the region of compression injury creates an environment allowing axonal sprouting that may be reversed with intraneural injections of purified myelin associated glycoprotein. These studies suggest that while the reciprocal relationship between neurons and glial cells is maintained, chronic nerve compression injury is a Schwann cell-mediated disease.
Collapse
Affiliation(s)
- Ranjan Gupta
- Department of Orthopaedic Surgery, Center for Biomedical Engineering in the Henry Samueli School of Engineering, University of California, Irvine, CA 92697, USA.
| | | | | |
Collapse
|
38
|
Bicker G. STOP and GO with NO: nitric oxide as a regulator of cell motility in simple brains. Bioessays 2005; 27:495-505. [PMID: 15832386 DOI: 10.1002/bies.20221] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
During the formation of the brain, neuronal cell migration and neurite extension are controlled by extracellular guidance cues. Here, I discuss experiments showing that the messenger nitric oxide (NO) is an additional regulator of cell motility. NO is a membrane permeant molecule, which activates soluble guanylyl cyclase (sGC) and leads to the formation of cyclic GMP (cGMP) in target cells. The analysis of specific cells types in invertebrate models such as molluscs, insects and the medicinal leech provides insight how NO and cyclic nucleotides affect the wiring of nervous systems by regulating cell and growth-cone motility. Inhibition of the NOS and sGC enzymes combined with rescue experiments show that NO signalling orchestrates neurite outgrowth and filopodial dynamics, cell migration of enteric neurons, glial migration and axonogenesis of pioneer fibers. Cultured insect embryos are accessible model systems in which cellular mechanisms of NO-induced cytoskeletal reorganizations can be analyzed in natural settings. Finally, I will outline some indications that NO may also regulate cell motility in the developing and regenerating vertebrate nervous system.
Collapse
Affiliation(s)
- Gerd Bicker
- School of Veterinary Medicine Hannover, Cell Biology, Institute of Physiology Bischofsholer Damm 15, D-30173 Hannover, Germany.
| |
Collapse
|
39
|
Park JW, Qi WN, Cai Y, Nunley JA, Urbaniak JR, Chen LE. The effects of exogenous nitric oxide donor on motor functional recovery of reperfused peripheral nerve. J Hand Surg Am 2005; 30:519-27. [PMID: 15925162 DOI: 10.1016/j.jhsa.2004.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2004] [Revised: 11/01/2004] [Accepted: 11/05/2004] [Indexed: 02/02/2023]
Abstract
PURPOSE To investigate the effects of the nitric oxide donor S-nitroso-N-acetylcysteine (SNAC) on motor functional recovery of reperfused rat sciatic nerve. METHODS Seventy-eight rats were divided into groups treated with SNAC (100 nmol/100 g/min), methylprednisolone 30 mg/kg/h for 15 minutes, 45-minute pause, 5.4 mg/kg/h for 1.5 h), and phosphate-buffered saline 0.2 mL/100 g/h). A 1-cm segment of sciatic nerve had 2 hours of ischemia and the results were evaluated after various reperfusion periods using a walking track test, muscle contractile testing, muscle weight, and histology. RESULTS During reperfusion there was a significant overall improvement in sciatic functional index measurement and isometric titanic contractile force for the SNAC-treated group compared with the methylprednisolone- and phosphate-buffered saline- treated groups. The SNAC group had significantly earlier improvement in the sciatic functional index measurement between days 7 and 28. Restoration of the contractile force and muscle weight of the extensor digitorum longus muscle began earlier in the SNAC group--after day 11--whereas the other 2 groups showed progressive atrophy until day 21, with a significant difference between the SNAC group and the other 2 groups. Histologic examination showed that SNAC-treated rats had less severe degeneration and earlier regeneration of axons than the others. Although methylprednisolone-treated rats showed earlier recovery than phosphate-buffered saline-treated rats in all parameters there were no significant differences between these 2 groups. CONCLUSIONS Supplementation of nitric oxide is effective in promoting motor functional recovery of the reperfused peripheral nerve and has potential to replace or augment steroids as therapeutic agents in treatment of nervous system ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jong Woong Park
- Department of Orthopaedic Surgery, College of Medicine, Korea University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
40
|
Revsin Y, Saravia F, Roig P, Lima A, de Kloet ER, Homo-Delarche F, De Nicola AF. Neuronal and astroglial alterations in the hippocampus of a mouse model for type 1 diabetes. Brain Res 2005; 1038:22-31. [PMID: 15748869 DOI: 10.1016/j.brainres.2004.12.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Revised: 12/14/2004] [Accepted: 12/17/2004] [Indexed: 11/23/2022]
Abstract
The influence of diabetes mellitus on brain pathology is increasingly recognized. Previous contributions of our laboratory demonstrated in models of type 1 diabetes (nonobese diabetic and streptozotocin (STZ)-treated mice), a marked astrogliosis and neurogenesis deficit in hippocampus and increased expression of hypothalamic neuropeptides. In the present investigation, we further analyzed alterations of astroglia and neurons in the hippocampus of mice 1 month after STZ-induced diabetes. Results showed that these STZ-diabetic mice presented: (a) increased number of astrocytes positive for apolipoprotein-E (Apo-E), a marker of ongoing neuronal dysfunction; (b) abnormal expression of early gene products associated with neuronal activation, including a high number of Jun + neurons in CA1 and CA3 layers and dentate gyrus, and of Fos-expressing neurons in CA3 layer; (c) augmented activity of NADPH-diaphorase, linked to oxidative stress, in CA3 region. These data support the concept that uncontrolled diabetes leads to hippocampal pathology, which adjoin to changes in other brain structures such as hypothalamus and cerebral cortex.
Collapse
Affiliation(s)
- Yanina Revsin
- Laboratory of Neuroendocrine Biochemistry, Institute of Biology and Experimental Medicine, Obligado 2490 (1428) Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
41
|
Keswani SC, Buldanlioglu U, Fischer A, Reed N, Polley M, Liang H, Zhou C, Jack C, Leitz GJ, Hoke A. A novel endogenous erythropoietin mediated pathway prevents axonal degeneration. Ann Neurol 2005; 56:815-26. [PMID: 15470751 DOI: 10.1002/ana.20285] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Clinically relevant peripheral neuropathies (such as diabetic and human immunodeficiency virus sensory neuropathies) are characterized by distal axonal degeneration, rather than neuronal death. Here, we describe a novel, endogenous pathway that prevents axonal degeneration. We show that in response to axonal injury, periaxonal Schwann cells release erythropoietin (EPO), which via EPO receptor binding on neurons, prevents axonal degeneration. We demonstrate that the relevant axonal injury signal that stimulates EPO production from surrounding glial cells is nitric oxide. In addition, we show that this endogenous pathway can be therapeutically exploited by administering exogenous EPO. In an animal model of distal axonopathy, systemic EPO administration prevents axonal degeneration, and this is associated with a reduction in limb weakness and neuropathic pain behavior. Our in vivo and in vitro data suggest that EPO prevents axonal degeneration and therefore may be therapeutically useful in a wide variety of human neurological diseases characterized by axonopathy.
Collapse
Affiliation(s)
- Sanjay C Keswani
- Department of Neurology, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gupta R, Gray M, Chao T, Bear D, Modafferi E, Mozaffar T. Schwann cells upregulate vascular endothelial growth factor secondary to chronic nerve compression injury. Muscle Nerve 2005; 31:452-60. [PMID: 15685607 DOI: 10.1002/mus.20272] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
To better understand the pathogenesis of chronic nerve compression injuries, we investigated the possibility that Schwann cell production of vascular endothelial growth factor (VEGF) is responsible for the increased vascularity and Schwann cell proliferation associated with chronic nerve injury. In situ hybridization was used to evaluate VEGF mRNA production with immunohistochemistry to further localize the production of VEGF and its receptor proteins in an animal model of chronic nerve compression injury. VEGF mRNA and protein expression increased within Schwann cells as early as 2 weeks after compression and peaked by 1 month with a subsequent marked increase in the number of blood vessels. Thus, chronic nerve compression injury induces Schwann cells to increase VEGF production, which may be responsible for changes in neural vasculature secondary to chronic nerve compression injury. With a better understanding of these nerve injuries, more effective treatments may be developed to help patients with these impairments.
Collapse
Affiliation(s)
- Ranjan Gupta
- Peripheral Nerve Research Laboratory, Department of Orthopedic Surgery, Medical Sciences I, Room B120, University of California, Irvine, California 92697, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Keilhoff G, Fansa H, Wolf G. Neuronal NOS deficiency promotes apoptotic cell death of spinal cord neurons after peripheral nerve transection. Nitric Oxide 2004; 10:101-11. [PMID: 15135363 DOI: 10.1016/j.niox.2004.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Revised: 03/23/2004] [Indexed: 12/11/2022]
Abstract
To study the role of endogenous NO in survival and recovery of spinal cord neurons after nerve lesions, wild type mice were compared to knock-out mice lacking neuronal, endothelial or inducible NO synthase (NOS) after sciatic nerve transection. The NO-generating capacities were assessed by NOS immunohistochemistry and NADPH-diaphorase staining. The feature of affected neurons was evaluated following Nissl- and TUNEL-staining, by immunocytochemical demonstration of cytochrome c-translocation, and by ultrastructural examination. Time point of cell loss was found to be independent of the mice type and occurred only at later post-axotomy states. The extent of neuronal degeneration, however, depended on the NO supply. Whereas a lack of endothelial or inducible NOS was well tolerated, deficiency of neuronal NOS enhanced the competence-to-die and led to a substantial apoptotic cell death of spinal cord neurons. Thus, NO supply turned out to be essential for cell survival and recovery with reference to the neuronal NOS isoform.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Medical Neurobiology, Otto-von-Guericke University, Leipziger Strasse 44, 39120 Magdeburg, Germany.
| | | | | |
Collapse
|
44
|
Gupta R, Lin YM, Bui P, Chao T, Preston C, Mozaffar T. Macrophage recruitment follows the pattern of inducible nitric oxide synthase expression in a model for carpal tunnel syndrome. J Neurotrauma 2003; 20:671-80. [PMID: 12908928 DOI: 10.1089/089771503322144581] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chronic nerve compression (CNC) induces a permeability change in neural vasculature. As recent evidence has shown that an alteration in reactive oxidative species (ROS) is related to neural degradation and regeneration, we evaluated whether inducible nitric oxide synthase (iNOS) plays a role in a rat model for CNC. Semi-quantitative analysis of iNOS mRNA and protein were performed with in situ hybridization and immunohistochemistry, respectively, at 3, 5, and 9 months post-operatively. At 3 months, iNOS mRNA was up-regulated in the perineurium of the proximal nerve with detectable changes in compressed and distal nerve segments. This expression continued to increase in the perineurium of 5-month proximal and compressed nerve segments with distal nerve demonstrating only a slight up-regulation of iNOS mRNA. At 9 months, iNOS mRNA expression was observed in both compressed and distal nerve. iNOS protein expression followed the same pattern of iNOS mRNA. As the perineurium is the blood-nerve barrier, the data suggests that these changes maybe mediated at the level of the perineurium. As macrophages release iNOS, we also evaluated whether macrophage recruitment followed the same pattern as iNOS expression. The results of ED-1 immunostaining for macrophages indicate that macrophages were localized to the outer one-third of cross sections during early time points. At later time points, macrophages were distributed diffusely throughout the nerve sections. Contrary to Wallerian degeneration, which elicits a relatively immediate signal for macrophage recruitment, CNC provides a slow, sustained stimulus for macrophage recruitment, which may be responsible for the up-regulation of iNOS gene expression.
Collapse
Affiliation(s)
- Ranjan Gupta
- Peripheral Nerve Research Lab, Department of Orthopaedic Surgery, University of California, Irvine, Med Sci I Room B120, Irvine, California 92697, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Keilhoff G, Fansa H, Wolf G. Neuronal nitric oxide synthase is the dominant nitric oxide supplier for the survival of dorsal root ganglia after peripheral nerve axotomy. J Chem Neuroanat 2002; 24:181-7. [PMID: 12297264 DOI: 10.1016/s0891-0618(02)00055-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
This study was designed to determine whether nitric oxide supply may be a major factor in the survival of dorsal root ganglia in a sciatic nerve injury model. Wild-type (WT) mice were compared with knockout (KO) mice lacking neuronal nitric oxide synthase (nNOS) or endothelial (eNOS). The NO-generating capacities were analysed by NOS immunohistochemistry and NADPH-diaphorase staining 1, 2, 6, and 12 weeks after nerve transection. The occurrence and morphological type of neuronal death were determined by TUNEL reaction and ultrastructural examination. Cell loss following nerve section, whist dependent on the availability of NO, as shown by its marked elevation in nNOS KO mice, did not correlate well with nNOS expression in WT animals. Whereas a lack of eNOS was tolerated, deficiency of nNOS led to an enhanced cell loss. The results suggest a crucial role of NO supply after transection of peripheral nerves with a particular significance of the nNOS isoform.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Medical Neurobiology, Medical Faculty, Otto-von-Guericke University, Leipziger Strasse 44, D-39120, Magdeburg, Germany.
| | | | | |
Collapse
|
46
|
Keilhoff G, Wolf G, Fansa H. NOS-mediated differences in peripheral nerve graft revascularization and regeneration. Neuroreport 2002; 13:1463-8. [PMID: 12167774 DOI: 10.1097/00001756-200208070-00023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Dependence of peripheral nerve revascularization on endogenous nitric oxide supply and consequences for nerve regeneration were investigated using a sciatic nerve graft model in mice lacking one of the nitric oxide synthase (NOS) isoforms. Wild type mice and mice lacking neuronal or inducible NOS exhibited similar revascularization patterns. Perfusion was consistently established 3 days after nerve reconstruction. Mice lacking endothelial NOS showed a delay in revascularization of about 2 days. The regeneration outcome did not reflect these differences. In mice lacking endothelial NOS, axon counts, myelination and recovery of sensory and motor function were comparable to wild type mice, whereas in mice lacking neuronal or inducible NOS a disturbed regeneration was found. Present results demonstrate, that the disturbance of nerve revascularization as result of reduced endothelial NOS-mediated NO supply can be tolerated, possibly by enhanced phagocytic capacity of Schwann cells and/or resident endoneurial macrophages.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Medical Neurobiology, Medical Faculty, Otto-von-Guericke-University, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | | | | |
Collapse
|