1
|
Doménech-Carbó A. Electrochemistry of plants: basic theoretical research and applications in plant science. J Solid State Electrochem 2021. [DOI: 10.1007/s10008-021-05046-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
2
|
Abstract
This study examines how the several major industries, associated with a carbon artifact production, essentially belong to one, closely knit family. The common parents are the geological fossils called petroleum and coal. The study also reviews the major developments in carbon nanotechnology and electrocatalysis over the last 30 years or so. In this context, the development of various carbon materials with size, dopants, shape, and structure designed to achieve high catalytic electroactivity is reported, and among them recent carbon electrodes with many important features are presented together with their relevant applications in chemical technology, neurochemical monitoring, electrode kinetics, direct carbon fuel cells, lithium ion batteries, electrochemical capacitors, and supercapattery.
Collapse
Affiliation(s)
- César A C Sequeira
- CeFEMA, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
- CeFEMA, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
| |
Collapse
|
3
|
Santhosh KT, Alizadeh A, Karimi-Abdolrezaee S. Design and optimization of PLGA microparticles for controlled and local delivery of Neuregulin-1 in traumatic spinal cord injury. J Control Release 2017; 261:147-162. [PMID: 28668379 DOI: 10.1016/j.jconrel.2017.06.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/21/2017] [Accepted: 06/27/2017] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) results in significant tissue damage that underlies functional impairments. Pharmacological interventions to confer neuroprotection and promote cell replacement are essential for SCI repair. We previously reported that Neuregulin-1 (Nrg-1) is acutely and permanently downregulated after SCI. Nrg-1 is a critical growth factor for differentiation of neural precursor cells (NPCs) into myelinating oligodendrocytes. We showed that intrathecal delivery of Nrg-1 enhances oligodendrocyte replacement following SCI. While an effective delivery system, intrathecal and systemic administration of growth factors with diverse biological targets may pose adverse off-target effects. Here, we have developed and optimized an injectable biodegradable poly(lactic-co-glycolic acid) (PLGA) microparticles system for sustained and prolonged intraspinal delivery of Nrg-1 in SCI. Recombinant human Nrg-1β1 peptide was encapsulated into PLGA microparticles. Optimal Nrg-1 release rate and duration were achieved by manipulating the porosity and size of PLGA particles. Our in vitro analysis showed a direct correlation between particle size and porosity with Nrg-1 release rate, while Nrg-1 loading efficiency in PLGA microparticles was inversely correlated with particle porosity. In SCI, local intraspinal injection of PLGA-Nrg-1 microparticles maintained significantly higher tissue levels of Nrg-1 for a long-term duration compared to Nrg-1 delivered intrathecally by osmotic pumps. Bioactivity of Nrg-1 in PLGA microparticles was verified by promoting oligodendrocyte differentiation of NPCs in vitro, and preservation of oligodendrocytes and axons in SCI. PLGA-Nrg-1 also attenuated neuroinflammation and glial scarring following SCI. We show, for the first time, the feasibility, efficacy and safety of PLGA microparticle system for local and controlled administration of Nrg-1 in SCI.
Collapse
Affiliation(s)
- Kallivalappil T Santhosh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
4
|
Xu Y, Zhang B, Messerli M, Randers-Pehrson G, Hei TK, Brenner DJ. Metabolic oxygen consumption measurement with a single-cell biosensor after particle microbeam irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2015; 54:137-144. [PMID: 25335641 PMCID: PMC4437628 DOI: 10.1007/s00411-014-0574-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 10/09/2014] [Indexed: 06/04/2023]
Abstract
A noninvasive, self-referencing biosensor/probe system has been integrated into the Columbia University Radiological Research Accelerator Facility Microbeam II end station. A single-cell oxygen consumption measurement has been conducted with this type of oxygen probe in 37° C Krebs-Ringer Bicarbonate buffer immediately before and after a single-cell microbeam irradiation. It is the first such measurement made for a microbeam irradiation, and a six fold increment of oxygen flux induced during a 15-s period of time has been observed following radiation exposure. The experimental procedure and the results are discussed.
Collapse
Affiliation(s)
- Yanping Xu
- Radiological Research Accelerator Facility (RARAF), Center for Radiological Research, Columbia University, 136 S. Broadway, Irvington, NY, 10533, USA.
| | - Bo Zhang
- Center for Radiological Research, Columbia University, New York, NY, 10032, USA
| | - Mark Messerli
- Biocurrents Research Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Gerhard Randers-Pehrson
- Radiological Research Accelerator Facility (RARAF), Center for Radiological Research, Columbia University, 136 S. Broadway, Irvington, NY, 10533, USA
| | - Tom K Hei
- Center for Radiological Research, Columbia University, New York, NY, 10032, USA
| | - David J Brenner
- Radiological Research Accelerator Facility (RARAF), Center for Radiological Research, Columbia University, 136 S. Broadway, Irvington, NY, 10533, USA
| |
Collapse
|
5
|
Innexin and pannexin channels and their signaling. FEBS Lett 2014; 588:1396-402. [PMID: 24632288 DOI: 10.1016/j.febslet.2014.03.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/06/2014] [Indexed: 01/24/2023]
Abstract
Innexins are bifunctional membrane proteins in invertebrates, forming gap junctions as well as non-junctional membrane channels (innexons). Their vertebrate analogues, the pannexins, have not only lost the ability to form gap junctions but are also prevented from it by glycosylation. Pannexins appear to form only non-junctional membrane channels (pannexons). The membrane channels formed by pannexins and innexins are similar in their biophysical and pharmacological properties. Innexons and pannexons are permeable to ATP, are present in glial cells, and are involved in activation of microglia by calcium waves in glia. Directional movement and accumulation of microglia following nerve injury, which has been studied in the leech which has unusually large glial cells, involves at least 3 signals: ATP is the "go" signal, NO is the "where" signal and arachidonic acid is a "stop" signal.
Collapse
|
6
|
The leech nervous system: a valuable model to study the microglia involvement in regenerative processes. Clin Dev Immunol 2013; 2013:274019. [PMID: 23878582 PMCID: PMC3710617 DOI: 10.1155/2013/274019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/07/2013] [Indexed: 11/25/2022]
Abstract
Microglia are intrinsic components of the central nervous system (CNS). During pathologies in mammals, inflammatory processes implicate the resident microglia and the infiltration of blood cells including macrophages. Functions of microglia appear to be complex as they exhibit both neuroprotective and neurotoxic effects during neuropathological conditions in vivo and in vitro. The medicinal leech Hirudo medicinalis is a well-known model in neurobiology due to its ability to naturally repair its CNS following injury. Considering the low infiltration of blood cells in this process, the leech CNS is studied to specify the activation mechanisms of only resident microglial cells. The microglia recruitment is known to be essential for the usual sprouting of injured axons and does not require any other glial cells. The present review will describe the questions which are addressed to understand the nerve repair. They will discuss the implication of leech factors in the microglial accumulation, the identification of nerve cells producing these molecules, and the study of different microglial subsets. Those questions aim to better understand the mechanisms of microglial cell recruitment and their crosstalk with damaged neurons. The study of this dialog is necessary to elucidate the balance of the inflammation leading to the leech CNS repair.
Collapse
|
7
|
Samuels SE, Lipitz JB, Wang J, Dahl G, Muller KJ. Arachidonic acid closes innexin/pannexin channels and thereby inhibits microglia cell movement to a nerve injury. Dev Neurobiol 2013; 73:621-31. [PMID: 23650255 DOI: 10.1002/dneu.22088] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/24/2013] [Accepted: 04/26/2013] [Indexed: 01/04/2023]
Abstract
Pannexons are membrane channels formed by pannexins and are permeable to ATP. They have been implicated in various physiological and pathophysiological processes. Innexins, the invertebrate homologues of the pannexins, form innexons. Nerve injury induces calcium waves in glial cells, releasing ATP through glial pannexon/innexon channels. The ATP then activates microglia. More slowly, injury releases arachidonic acid (ArA). The present experiments show that ArA itself reduced the macroscopic membrane currents of innexin- and of pannexin-injected oocytes; ArA also blocked K(+) -induced release of ATP. In leeches, whose large glial cells have been favorable for studying control of microglia migration, ArA blocked glial dye-release and, evidently, ATP-release. A physiological consequence in the leech was block of microglial migration to nerve injuries. Exogenous ATP (100 µM) reversed the effect, for ATP causes activation and movement of microglia after nerve injury, but nitric oxide directs microglia to the lesion. It was not excluded that metabolites of ArA may also inhibit the channels. But for all these effects, ArA and its non-metabolizable analog eicosatetraynoic acid (ETYA) were indistinguishable. Therefore, ArA itself is an endogenous regulator of pannexons and innexons. ArA thus blocks release of ATP from glia after nerve injury and thereby, at least in leeches, stops microglia at lesions.
Collapse
Affiliation(s)
- Stuart E Samuels
- Neuroscience Program, University of Miami, Miami, Florida, 33136, USA
| | | | | | | | | |
Collapse
|
8
|
Arafah K, Croix D, Vizioli J, Desmons A, Fournier I, Salzet M. Involvement of nitric oxide through endocannabinoids release in microglia activation during the course of CNS regeneration in the medicinal leech. Glia 2013; 61:636-49. [DOI: 10.1002/glia.22462] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 12/17/2012] [Indexed: 11/06/2022]
|
9
|
Tahtouh M, Garçon-Bocquet A, Croq F, Vizioli J, Sautière PE, Van Camp C, Salzet M, Nagnan-le Meillour P, Pestel J, Lefebvre C. Interaction of HmC1q with leech microglial cells: involvement of C1qBP-related molecule in the induction of cell chemotaxis. J Neuroinflammation 2012; 9:37. [PMID: 22356764 PMCID: PMC3298539 DOI: 10.1186/1742-2094-9-37] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 02/22/2012] [Indexed: 01/24/2023] Open
Abstract
Background In invertebrates, the medicinal leech is considered to be an interesting and appropriate model to study neuroimmune mechanisms. Indeed, this non-vertebrate animal can restore normal function of its central nervous system (CNS) after injury. Microglia accumulation at the damage site has been shown to be required for axon sprouting and for efficient regeneration. We characterized HmC1q as a novel chemotactic factor for leech microglial cell recruitment. In mammals, a C1q-binding protein (C1qBP alias gC1qR), which interacts with the globular head of C1q, has been reported to participate in C1q-mediated chemotaxis of blood immune cells. In this study, we evaluated the chemotactic activities of a recombinant form of HmC1q and its interaction with a newly characterized leech C1qBP that acts as its potential ligand. Methods Recombinant HmC1q (rHmC1q) was produced in the yeast Pichia pastoris. Chemotaxis assays were performed to investigate rHmC1q-dependent microglia migration. The involvement of a C1qBP-related molecule in this chemotaxis mechanism was assessed by flow cytometry and with affinity purification experiments. The cellular localization of C1qBP mRNA and protein in leech was investigated using immunohistochemistry and in situ hybridization techniques. Results rHmC1q-stimulated microglia migrate in a dose-dependent manner. This rHmC1q-induced chemotaxis was reduced when cells were preincubated with either anti-HmC1q or anti-human C1qBP antibodies. A C1qBP-related molecule was characterized in leech microglia. Conclusions A previous study showed that recruitment of microglia is observed after HmC1q release at the cut end of axons. Here, we demonstrate that rHmC1q-dependent chemotaxis might be driven via a HmC1q-binding protein located on the microglial cell surface. Taken together, these results highlight the importance of the interaction between C1q and C1qBP in microglial activation leading to nerve repair in the medicinal leech.
Collapse
Affiliation(s)
- Muriel Tahtouh
- Laboratoire de Spectrométrie de Masse Biologique Fondamentale et Appliquée - EA4550, Microglial activation group, Université Lille Nord de France, Université Lille 1, IFR 147, bâtiment SN3, 59655 Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Microglial cells are the resident macrophages in the central nervous system. These cells of mesodermal/mesenchymal origin migrate into all regions of the central nervous system, disseminate through the brain parenchyma, and acquire a specific ramified morphological phenotype termed "resting microglia." Recent studies indicate that even in the normal brain, microglia have highly motile processes by which they scan their territorial domains. By a large number of signaling pathways they can communicate with macroglial cells and neurons and with cells of the immune system. Likewise, microglial cells express receptors classically described for brain-specific communication such as neurotransmitter receptors and those first discovered as immune cell-specific such as for cytokines. Microglial cells are considered the most susceptible sensors of brain pathology. Upon any detection of signs for brain lesions or nervous system dysfunction, microglial cells undergo a complex, multistage activation process that converts them into the "activated microglial cell." This cell form has the capacity to release a large number of substances that can act detrimental or beneficial for the surrounding cells. Activated microglial cells can migrate to the site of injury, proliferate, and phagocytose cells and cellular compartments.
Collapse
|
11
|
Croq F, Vizioli J, Tuzova M, Tahtouh M, Sautiere PE, Van Camp C, Salzet M, Cruikshank WW, Pestel J, Lefebvre C. A homologous form of human interleukin 16 is implicated in microglia recruitment following nervous system injury in leech Hirudo medicinalis. Glia 2011; 58:1649-62. [PMID: 20578037 DOI: 10.1002/glia.21036] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In contrast to mammals, the medicinal leech Hirudo medicinalis can completely repair its central nervous system (CNS) after injury. This invertebrate model offers unique opportunities to study the molecular and cellular basis of the CNS repair processes. When the leech CNS is injured, microglial cells migrate and accumulate at the site of lesion, a phenomenon known to be essential for the usual sprouting of injured axons. In the present study, we demonstrate that a new molecule, designated HmIL-16, having functional homologies with human interleukin-16 (IL-16), has chemotactic activity on leech microglial cells as observed using a gradient of human IL-16. Preincubation of microglial cells either with an anti-human IL-16 antibody or with anti-HmIL-16 antibody significantly reduced microglia migration induced by leech-conditioned medium. Functional homology was demonstrated further by the ability of HmIL-16 to promote human CD4+ T cell migration which was inhibited by antibody against human IL-16, an IL-16 antagonist peptide or soluble CD4. Immunohistochemistry of leech CNS indicates that HmIL-16 protein present in the neurons is rapidly transported and stored along the axonal processes to promote the recruitment of microglial cells to the injured axons. To our knowledge, this is the first identification of a functional interleukin-16 homologue in invertebrate CNS. The ability of HmIL-16 to recruit microglial cells to sites of CNS injury suggests a role for HmIL-16 in the crosstalk between neurons and microglia in the leech CNS repair.
Collapse
Affiliation(s)
- Françoise Croq
- Université Lille Nord de France, Université Lille 1, Laboratoire de Neuroimmunologie des Annélides, Centre National de la Recherche Scientifique, FRE 3249, IFR 147, F59655 Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Samuels SE, Lipitz JB, Dahl G, Muller KJ. Neuroglial ATP release through innexin channels controls microglial cell movement to a nerve injury. ACTA ACUST UNITED AC 2011; 136:425-42. [PMID: 20876360 PMCID: PMC2947054 DOI: 10.1085/jgp.201010476] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Microglia, the immune cells of the central nervous system, are attracted to sites of injury. The injury releases adenosine triphosphate (ATP) into the extracellular space, activating the microglia, but the full mechanism of release is not known. In glial cells, a family of physiologically regulated unpaired gap junction channels called innexons (invertebrates) or pannexons (vertebrates) located in the cell membrane is permeable to ATP. Innexons, but not pannexons, also pair to make gap junctions. Glial calcium waves, triggered by injury or mechanical stimulation, open pannexon/innexon channels and cause the release of ATP. It has been hypothesized that a glial calcium wave that triggers the release of ATP causes rapid microglial migration to distant lesions. In the present study in the leech, in which a single giant glial cell ensheathes each connective, hydrolysis of ATP with 10 U/ml apyrase or block of innexons with 10 µM carbenoxolone (CBX), which decreased injury-induced ATP release, reduced both movement of microglia and their accumulation at lesions. Directed movement and accumulation were restored in CBX by adding ATP, consistent with separate actions of ATP and nitric oxide, which is required for directed movement but does not activate glia. Injection of glia with innexin2 (Hminx2) RNAi inhibited release of carboxyfluorescein dye and microglial migration, whereas injection of innexin1 (Hminx1) RNAi did not when measured 2 days after injection, indicating that glial cells’ ATP release through innexons was required for microglial migration after nerve injury. Focal stimulation either mechanically or with ATP generated a calcium wave in the glial cell; injury caused a large, persistent intracellular calcium response. Neither the calcium wave nor the persistent response required ATP or its release. Thus, in the leech, innexin membrane channels releasing ATP from glia are required for migration and accumulation of microglia after nerve injury.
Collapse
Affiliation(s)
- Stuart E Samuels
- Neuroscience Program, University of Miami School of Medicine, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
13
|
McLamore ES, Porterfield DM. Non-invasive tools for measuring metabolism and biophysical analyte transport: self-referencing physiological sensing. Chem Soc Rev 2011; 40:5308-20. [DOI: 10.1039/c0cs00173b] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Lv P, Rodriguez-Contreras A, Kim HJ, Zhu J, Wei D, Choong-Ryoul S, Eastwood E, Mu K, Levic S, Song H, Yevgeniy PY, Smith PJS, Yamoah EN. Release and elementary mechanisms of nitric oxide in hair cells. J Neurophysiol 2010; 103:2494-505. [PMID: 20220083 DOI: 10.1152/jn.00017.2010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The enzyme nitric oxide (NO) synthase, that produces the signaling molecule NO, has been identified in several cell types in the inner ear. However, it is unclear whether a measurable quantity of NO is released in the inner ear to confer specific functions. Indeed, the functional significance of NO and the elementary cellular mechanism thereof are most uncertain. Here, we demonstrate that the sensory epithelia of the frog saccule release NO and explore its release mechanisms by using self-referencing NO-selective electrodes. Additionally, we investigated the functional effects of NO on electrical properties of hair cells and determined their underlying cellular mechanism. We show detectable amounts of NO are released by hair cells (>50 nM). Furthermore, a hair-cell efferent modulator acetylcholine produces at least a threefold increase in NO release. NO not only attenuated the baseline membrane oscillations but it also increased the magnitude of current required to generate the characteristic membrane potential oscillations. This resulted in a rightward shift in the frequency-current relationship and altered the excitability of hair cells. Our data suggest that these effects ensue because NO reduces whole cell Ca(2+) current and drastically decreases the open probability of single-channel events of the L-type and non L-type Ca(2+) channels in hair cells, an effect that is mediated through direct nitrosylation of the channel and activation of protein kinase G. Finally, NO increases the magnitude of Ca(2+)-activated K(+) currents via direct NO nitrosylation. We conclude that NO-mediated inhibition serves as a component of efferent nerve modulation of hair cells.
Collapse
Affiliation(s)
- Ping Lv
- Center for Neuroscience, Department of Anesthesiology and Pain Medicine, Program in Communication Sciences, University of California, Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Duan Y, Sahley CL, Muller KJ. ATP and NO dually control migration of microglia to nerve lesions. Dev Neurobiol 2009; 69:60-72. [PMID: 19025930 DOI: 10.1002/dneu.20689] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microglia migrate rapidly to lesions in the central nervous system (CNS), presumably in response to chemoattractants including ATP released directly or indirectly by the injury. Previous work on the leech has shown that nitric oxide (NO), generated at the lesion, is both a stop signal for microglia at the lesion and crucial for their directed migration from hundreds of micrometers away within the nerve cord, perhaps mediated by a soluble guanylate cyclase (sGC). In this study, application of 100 microM ATP caused maximal movement of microglia in leech nerve cords. The nucleotides ADP, UTP, and the nonhydrolyzable ATP analog AMP-PNP (adenyl-5'-yl imidodiphosphate) also caused movement, whereas AMP, cAMP, and adenosine were without effect. Both movement in ATP and migration after injury were slowed by 50 microM reactive blue 2 (RB2), an antagonist of purinergic receptors, without influencing the direction of movement. This contrasted with the effect of the NO scavenger cPTIO (2-(4-carboxyphenyl)-4,4,5,5-teramethylimidazoline-oxyl-3-oxide), which misdirected movement when applied at 1 mM. The cPTIO reduced cGMP immunoreactivity without changing the immunoreactivity of eNOS (endothelial nitric oxide synthase), which accompanies increased NOS activity after nerve cord injury, consistent with involvement of sGC. Moreover, the sGC-specific inhibitor LY83583 applied at 50 microM had a similar effect, in agreement with previous results with methylene blue. Taken together, the experiments support the hypothesis that ATP released directly or indirectly by injury activates microglia to move, whereas NO that activates sGC directs migration of microglia to CNS lesions.
Collapse
Affiliation(s)
- Yuanli Duan
- Department of Physiology and Biophysics (R-430), University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | |
Collapse
|
16
|
Bigelow A, Garty G, Funayama T, Randers-Pehrson G, Brenner D, Geard C. Expanding the question-answering potential of single-cell microbeams at RARAF, USA. JOURNAL OF RADIATION RESEARCH 2009; 50 Suppl A:A21-8. [PMID: 19346682 PMCID: PMC2924733 DOI: 10.1269/jrr.08134s] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Charged-particle microbeams, developed to provide targeted irradiation of individual cells, and then of sub-cellular components, and then of 3-D tissues and now organisms, have been instrumental in challenging and changing long accepted paradigms of radiation action. However the potential of these valuable tools can be enhanced by integrating additional components with the direct ability to measure biological responses in real time, or to manipulate the cell, tissue or organism of interest under conditions where information gained can be optimized. The RARAF microbeam has recently undergone an accelerator upgrade, and been modified to allow for multiple microbeam irradiation laboratories. Researchers with divergent interests have expressed desires for particular modalities to be made available and ongoing developments reflect these desires. The focus of this review is on the design, incorporation and use of multiphoton and other imaging, micro-manipulation and single cell biosensor capabilities at RARAF. Additionally, an update on the status of the other biology oriented microbeams in the Americas is provided.
Collapse
Affiliation(s)
- Alan Bigelow
- Center for Radiological Research, Columbia University, Irvington, New York 10533-0021, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Tahtouh M, Croq F, Vizioli J, Sautiere PE, Van Camp C, Salzet M, Daha MR, Pestel J, Lefebvre C. Evidence for a novel chemotactic C1q domain-containing factor in the leech nerve cord. Mol Immunol 2009; 46:523-31. [DOI: 10.1016/j.molimm.2008.07.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 07/15/2008] [Accepted: 07/15/2008] [Indexed: 10/21/2022]
|
18
|
Koehler JJ, Zhao J, Jedlicka SS, Porterfield DM, Rickus JL. Compartmentalized Nanocomposite for Dynamic Nitric Oxide Release. J Phys Chem B 2008; 112:15086-93. [DOI: 10.1021/jp803276u] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- John J. Koehler
- Department of Agricultural and Biological Engineering, Department of Horticulture and Landscape Architecture, Weldon School of Biomedical Engineering, Physiological Sensing Facility at the Bindley Bioscience Center, Purdue University, West Lafayette, IN
| | - Jianxiu Zhao
- Department of Agricultural and Biological Engineering, Department of Horticulture and Landscape Architecture, Weldon School of Biomedical Engineering, Physiological Sensing Facility at the Bindley Bioscience Center, Purdue University, West Lafayette, IN
| | - Sabrina S. Jedlicka
- Department of Agricultural and Biological Engineering, Department of Horticulture and Landscape Architecture, Weldon School of Biomedical Engineering, Physiological Sensing Facility at the Bindley Bioscience Center, Purdue University, West Lafayette, IN
| | - D. Marshall Porterfield
- Department of Agricultural and Biological Engineering, Department of Horticulture and Landscape Architecture, Weldon School of Biomedical Engineering, Physiological Sensing Facility at the Bindley Bioscience Center, Purdue University, West Lafayette, IN
| | - Jenna L. Rickus
- Department of Agricultural and Biological Engineering, Department of Horticulture and Landscape Architecture, Weldon School of Biomedical Engineering, Physiological Sensing Facility at the Bindley Bioscience Center, Purdue University, West Lafayette, IN
| |
Collapse
|
19
|
Martinez VG, Manson JM, Zoran MJ. Effects of nerve injury and segmental regeneration on the cellular correlates of neural morphallaxis. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2008; 310:520-33. [PMID: 18561185 PMCID: PMC2754161 DOI: 10.1002/jez.b.21224] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Functional recovery of neural networks after injury requires a series of signaling events similar to the embryonic processes that governed initial network construction. Neural morphallaxis, a form of nervous system regeneration, involves reorganization of adult neural connectivity patterns. Neural morphallaxis in the worm, Lumbriculus variegatus, occurs during asexual reproduction and segmental regeneration, as body fragments acquire new positional identities along the anterior-posterior axis. Ectopic head (EH) formation, induced by ventral nerve cord lesion, generated morphallactic plasticity including the reorganization of interneuronal sensory fields and the induction of a molecular marker of neural morphallaxis. Morphallactic changes occurred only in segments posterior to an EH. Neither EH formation, nor neural morphallaxis was observed after dorsal body lesions, indicating a role for nerve cord injury in morphallaxis induction. Furthermore, a hierarchical system of neurobehavioral control was observed, where anterior heads were dominant and an EH controlled body movements only in the absence of the anterior head. Both suppression of segmental regeneration and blockade of asexual fission, after treatment with boric acid, disrupted the maintenance of neural morphallaxis, but did not block its induction. Therefore, segmental regeneration (i.e., epimorphosis) may not be required for the induction of morphallactic remodeling of neural networks. However, on-going epimorphosis appears necessary for the long-term consolidation of cellular and molecular mechanisms underlying the morphallaxis of neural circuitry.
Collapse
Affiliation(s)
| | | | - Mark J. Zoran
- Department of Biology, Texas A&M University, College Station, Texas
| |
Collapse
|
20
|
van der Zeyden M, Oldenziel WH, Rea K, Cremers TI, Westerink BH. Microdialysis of GABA and glutamate: analysis, interpretation and comparison with microsensors. Pharmacol Biochem Behav 2007; 90:135-47. [PMID: 17939932 DOI: 10.1016/j.pbb.2007.09.004] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 08/16/2007] [Accepted: 09/04/2007] [Indexed: 10/22/2022]
Abstract
GABA and glutamate sampled from the brain by microdialysis do not always fulfill the classic criteria for exocytotic release. In this regard the origin (neuronal vs. astroglial, synaptic vs. extrasynaptic) of glutamate and GABA collected by microdialysis as well as in the ECF itself, is still a matter of debate. In this overview microdialysis of GABA and glutamate and the use of microsensors to detect extracellular glutamate are compared and discussed. During basal conditions glutamate in microdialysates is mainly derived from non-synaptic sources. Indeed recently several sources of astrocytic glutamate release have been described, including glutamate derived from gliotransmission. However during conditions of (chemical, electrical or behavioral) stimulation a significant part of glutamate might be derived from neurotransmission. Interestingly accumulating evidence suggests that glutamate determined by microsensors is more likely to reflect basal synaptic events. This would mean that microdialysis and microsensors are complementary methods to study extracellular glutamate. Regarding GABA we concluded that the chromatographic conditions for the separation of this transmitter from other amino acid-derivatives are extremely critical. Optimal conditions to detect GABA in microdialysis samples--at least in our laboratory--include a retention time of approximately 60 min and a careful control of the pH of the mobile phase. Under these conditions it appears that 50-70% of GABA in dialysates is derived from neurotransmission.
Collapse
Affiliation(s)
- Miranda van der Zeyden
- Department of Biomonitoring and Sensoring, University Centre for Pharmacy, Antonius Deusinglaan 1, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
21
|
Porterfield DM. Measuring metabolism and biophysical flux in the tissue, cellular and sub-cellular domains: Recent developments in self-referencing amperometry for physiological sensing. Biosens Bioelectron 2007; 22:1186-96. [PMID: 16870420 DOI: 10.1016/j.bios.2006.06.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Revised: 05/24/2006] [Accepted: 06/05/2006] [Indexed: 11/23/2022]
Abstract
Ultimately, advances in genomics, proteomics and metabolomics will be realized by combining these approaches with biophysical sensors for understanding the functional and structural (physiological) aspects of sub-cellular systems (cytomics). Therefore, the emergence of the new fields of cytomics and physiomics will require new technologies to probe the functional realm of living cells. While amperometric sensors have been used, their sensitivity and reliability are significantly improved through the development of new strategies and data acquisition systems for the operation of the sensors. This includes the application of the principles of the vibrating or self-referencing microsensor to the operation of amperometric sensors. The development of self-referencing amperometry (SRA) is significant because it effectively converts static concentration sensors into dynamic biophysical sensors that directly monitor physiological flux. SRA has been developed for analytes such as O2, NO, H2O2 and ascorbate. These sensors have been validated against non-biological microscopic flux sources that were theoretically modeled, before being applied to biological research. This new sensor technology has been shown, through research in a wide variety of biological and biomedical research projects, to be an important new tool in the arsenal of the cell biologist. SRA technology has been adapted through SRA-H2O2 and SRA-NADH sensors, for electrochemically coupled enzyme based self-referencing biosensors (SRB) for glucose, glutamate and ethanol. These developments in self-referencing sensor technologies offer great promise in extending electroanalytical chemistry and biosensor technologies from the micro to the nanoscale where researchers can study physiology at the sub-cellular and organellar levels.
Collapse
Affiliation(s)
- D Marshall Porterfield
- Department of Agricultural and Biological Engineering, Bindley Bioscience Center: Physiological Sensing Facility, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, United States
| |
Collapse
|
22
|
Yahyavi-Firouz-Abadi N, Tahsili-Fahadan P, Ostad SN. Effect of μ and κ opioids on injury-induced microglial accumulation in leech CNS: Involvement of the nitric oxide pathway. Neuroscience 2007; 144:1075-86. [PMID: 17169497 DOI: 10.1016/j.neuroscience.2006.10.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2006] [Revised: 10/22/2006] [Accepted: 10/26/2006] [Indexed: 11/30/2022]
Abstract
Damage to the leech or mammalian CNS increases nitric oxide (NO) production and causes accumulation of phagocytic microglial cells at the injury site. Opioids have been postulated to modulate various parameters of the immune response. Morphine and leech morphine-like substance are shown to release NO and suppress microglial activation. Regarding the known immuno-modulatory effects of selective mu and kappa ligands, we have assessed the effect of these agents on accumulation of microglia at the site of injury in leech CNS. Leech nerve cords were dissected, crushed with fine forceps and maintained in different concentrations of opiates in culture medium for 3 h and then fixed and double stained with Hoechst 33258 and monoclonal antibody to endothelial nitric oxide synthase (NOS). Morphine and naloxone (> or =10(-3) M) but not selective mu agonist, DAMGO [d-Ala2, N-Me-Phe-Gly5(ol)-enkephalin] and antagonist, CTAP [D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2] inhibited the microglial accumulation. The effect of morphine was abrogated by pre-treatment with naloxone and also non-selective NOS inhibitor, l-NAME [N(omega)-nitro-l-arginine-methyl-ester; 10(-3) M] implying an NO-dependent and mu-mediated mechanism. These results are similar to properties of recently found mu-3 receptor in leech, which is sensitive to alkaloids but not peptides. Both selective kappa agonist, U50,488 [3,4-dichloro-N-methyl-N-(2-(1-pyrrolidinyl)cyclohexyl)-benzeneacetamide; > or =10(-3) M], and antagonist, nor-binaltorphimine (nor-BNI; > or =10(-3) M), inhibited the accumulation. The effect of nor-BNI was reversed by l-NAME. Immunohistochemistry showed decreased endothelial NOS expression in naloxone and U50,488-treated cords. Since, NO production at the injury site is hypothesized to act as a stop signal for microglias, opioid agents may exert their effect via changing of NO gradient along the cord resulting in disruption of accumulation. These results suggest an immuno-modulatory role for mu and kappa opioid receptors on injury-induced microglial accumulation which may be mediated via NO.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Enzyme Inhibitors/pharmacology
- Gliosis/metabolism
- Gliosis/physiopathology
- Hirudo medicinalis/cytology
- Hirudo medicinalis/metabolism
- Microglia/cytology
- Microglia/metabolism
- NG-Nitroarginine Methyl Ester/pharmacology
- Narcotic Antagonists/pharmacology
- Nervous System/cytology
- Nervous System/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/metabolism
- Opioid Peptides/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Trauma, Nervous System/metabolism
- Trauma, Nervous System/physiopathology
Collapse
Affiliation(s)
- N Yahyavi-Firouz-Abadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Enghelab Avenue, P.O. Box 14155/6451, Tehran, Iran
| | | | | |
Collapse
|
23
|
Babington EJ, Vatanparast J, Verrall J, Blackshaw SE. Three-dimensional culture of leech and snail ganglia for studies of neural repair. INVERTEBRATE NEUROSCIENCE 2005; 5:173-82. [PMID: 16172883 DOI: 10.1007/s10158-005-0006-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Accepted: 08/04/2005] [Indexed: 10/25/2022]
Abstract
Three-dimensional (3D) collagen gels provide a stable matrix in which isolated regenerating ganglia from leech and snail can be maintained for studies of the molecular and cellular mechanisms underlying the regenerative process. Segmental ganglia from leech, or supraoesophageal, suboesophageal or buccal ganglia from snail were maintained for up to 3 weeks in 3D matrices of mammalian Type I collagen. The collagen matrix supports the regenerative outgrowth of axon tracts as well as the migration of microglial cells, important elements in the repair process. Proteins or soluble factors or target tissue may be added to the basic collagen matrix to manipulate the environment of the regenerating tissue. We describe techniques for immunostaining of regenerating axons and microglial cells within the gel matrix in combination with staining of cell nuclei, and the use of intracellular labelling to distinguish axons of identified neurons within the regenerative outgrowth.
Collapse
Affiliation(s)
- E J Babington
- Department of Human Anatomy & Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | | | | | | |
Collapse
|
24
|
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
|
25
|
Duan Y, Panoff J, Burrell BD, Sahley CL, Muller KJ. Repair and regeneration of functional synaptic connections: cellular and molecular interactions in the leech. Cell Mol Neurobiol 2005; 25:441-50. [PMID: 16047551 DOI: 10.1007/s10571-005-3152-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A major problem for neuroscience has been to find a means to achieve reliable regeneration of synaptic connections following injury to the adult CNS. This problem has been solved by the leech, where identified neurons reconnect precisely with their usual targets following axotomy, re-establishing in the adult the connections formed during embryonic development. It cannot be assumed that once axons regenerate specific synapses, function will be restored. Recent work on the leech has shown following regeneration of the synapse between S-interneurons, which are required for sensitization of reflexive shortening, a form of non-associative learning, the capacity for sensitization is delayed. The steps in repair of synaptic connections in the leech are reviewed, with the aim of understanding general mechanisms that promote successful repair. New results are presented regarding the signals that regulate microglial migration to lesions, a first step in the repair process. In particular, microglia up to 900 microm from the lesion respond within minutes by moving rapidly toward the injury, controlled in part by nitric oxide (NO), which is generated immediately at the lesion and acts via a soluble guanylate cyclase (sGC). The cGMP produced remains elevated for hours after injury. The relationship of microglial migration to axon outgrowth is discussed.
Collapse
Affiliation(s)
- Yuanli Duan
- Department of Physiology & Biophysics, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | |
Collapse
|
26
|
Grassi C, D'Ascenzo M, Azzena GB. Modulation of Ca(v)1 and Ca(v)2.2 channels induced by nitric oxide via cGMP-dependent protein kinase. Neurochem Int 2004; 45:885-93. [PMID: 15312983 DOI: 10.1016/j.neuint.2004.03.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The unconventional gaseous transmitter nitric oxide (NO) markedly influences most of mechanisms involved in the regulation of intracellular Ca2+ homeostasis. In excitable cells, Ca2+ signaling mainly depends on the activity of voltage-gated Ca2+ channels (VGCCs). In the present paper, we will review data from our laboratory and others characterizing NO-induced modulation of Ca(v)1 (L-type) and Ca(v)2.2 (N-type) channels. In particular, we will explore experimental evidence indicating that NO's inhibition of channel gating is produced via cGMP-dependent protein kinase and examine some of the numerous cell functions that are potentially influenced by the action of NO on Ca2+ channels.
Collapse
Affiliation(s)
- Claudio Grassi
- Institute of Human Physiology, Medical School, Catholic University S. Cuore, Largo F. Vito 1, I-00168 Rome, Italy.
| | | | | |
Collapse
|
27
|
Piao HZ, Jin SA, Chun HS, Lee JC, Kim WK. Neuroprotective effect of wogonin: Potential roles of inflammatory cytokines. Arch Pharm Res 2004; 27:930-6. [PMID: 15473663 DOI: 10.1007/bf02975846] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Wogonin (5,7-dihydroxy-8-methoxyflavone), an active component originated from the root of Scutellaria baicalensis Georgi, has been reported to possess antioxidant and anti-inflammatory properties. In this study, we investigated the neuroprotective effect of wogonin in a focal cerebral ischemia rat model. Wogonin markedly reduced the infarct volume after 2 h middle cerebral artery occlusion followed by 22 h reperfusion. Wogonin decreased the production of nitric oxide and inflammatory cytokines such as TNF-alpha and IL-6 in lipopolisaccharide-stimulated microglial cells. While wogonin reduced the activity of NF-kappaB, it did not change the activity of mitogen-activated protein kinases family members, p38, ERK and JNK. The lipopolisaccharide-stimulated production of NO and cytokines was significantly blocked by various kinds of NF-kappaB inhibitors such as N-acetyl cysteine, pyrrolidinedithiocarbamate and MG-132. The data may indicate that wogonin has neuroprotective effect by preventing the overactivation of microglial cells, possibly by inactivating NF-kappaB signaling pathway.
Collapse
Affiliation(s)
- Hua Zi Piao
- Department of Pharmacology, College of Medicine, Ewha Institute of Neuroscience, Ewha Womens University, Seoul, Korea
| | | | | | | | | |
Collapse
|
28
|
Peruzzi E, Fontana G, Sonetti D. Presence and role of nitric oxide in the central nervous system of the freshwater snail Planorbarius corneus: possible implication in neuron–microglia communication. Brain Res 2004; 1005:9-20. [PMID: 15044059 DOI: 10.1016/j.brainres.2003.12.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2003] [Indexed: 11/19/2022]
Abstract
The aim of the present study was to investigate the involvement of nitric oxide (NO) as a messenger molecule in neuron-microglia communication in the central nervous system (CNS) of the freshwater snail Planorbarius corneus. The presence of both neuronal (nNOS) and inducible nitric oxide synthase (iNOS) was studied using NADPH-diaphorase (NADPH-d) histochemistry and NOS immunocytochemistry. The experiments were performed on whole ganglia and cultured microglial cells after different activation modalities, such as treatment with lipopolysaccharide and adenosine triphosphate and/or maintaining ganglia in culture medium till 7 days. In sections, nNOS immunoreactivity was found only in neurons and nNOS-positive elements were less numerous than NADPH-d-positive ones, with which they partially overlapped. The iNOS immunoreactivity was observed only after activation, in both nerve and microglial cells. We also found that the number of iNOS-immunoreactive neurons and microglia varied, depending on the activation modalities. In microglial cell cultures, iNOS was expressed in the first generation of cells only after activation, whereas a second generation, proliferated after ganglia activation, expressed iNOS even in the unstimulated condition.
Collapse
Affiliation(s)
- Elisa Peruzzi
- Department of Animal Biology, University of Modena and Reggio Emilia, Via Campi 213/D, I-41100 Modena, Italy.
| | | | | |
Collapse
|
29
|
Duan Y, Haugabook SJ, Sahley CL, Muller KJ. Methylene blue blocks cGMP production and disrupts directed migration of microglia to nerve lesions in the leech CNS. ACTA ACUST UNITED AC 2003; 57:183-92. [PMID: 14556284 DOI: 10.1002/neu.10262] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Migration and accumulation of microglial cells at sites of injury are important for nerve repair. Recent studies on the leech central nervous system (CNS), in which synapse regeneration is successful, have shown that nitric oxide (NO) generated immediately after injury by endothelial nitric oxide synthase (eNOS) stops migrating microglia at the lesion. The present study obtained results indicating that NO may act earlier, on microglia migration, and aimed to determine mechanisms underlying NO's effects. Injury induced cGMP immunoreactivity at the lesion in a pattern similar to that of eNOS activity, immunoreactivity, and microglial cell accumulation, which were all focused there. The soluble guanylate cyclase (sGC) inhibitor methylene blue (MB) at 60 microM abolished cGMP immunoreactivity at lesions and blocked microglial cell migration and accumulation without interfering with axon conduction. Time-lapse video microscopy of microglia in living nerve cords showed MB did not reduce cell movement but reduced directed movement, with significantly more cells moving away from the lesion or reversing direction and fewer cells moving toward the lesion. The results indicate a new role for NO, directing the microglial cell migration as well as stopping it, and show that NO's action may be mediated by cGMP.
Collapse
Affiliation(s)
- Yuanli Duan
- Department of Physiology and Biophysics (R-430), University of Miami School of Medicine, 1600 NW 10th Avenue, Miami, Florida 33136, USA.
| | | | | | | |
Collapse
|
30
|
Abstract
Nitric oxide (NO) amperometric microsensor was prepared by the modification of bare carbon fiber electrode by Nafion and cellulose acetate (CA). Detection limit, response time, reproducibility and influence of some possible interferences (nitrite, nitrate, arginine) were tested and evaluated. This sensor was used for in vitro determination of NO release from fresh porcine aorta induced by calcium ionophore A23187 (CI).
Collapse
Affiliation(s)
- Jaroslav Katrlík
- Department of Analytical Chemistry, Faculty of Pharmacy, Comenius University, Odbojárov 10, SK-832 32 Bratislava, Slovakia.
| | | |
Collapse
|
31
|
Smith P, Haydon P, Hengstenberg A, Jung SK. Analysis of cellular boundary layers: application of electrochemical microsensors. Electrochim Acta 2001. [DOI: 10.1016/s0013-4686(01)00567-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Jung SK, Trimarchi JR, Sanger RH, Smith PJ. Development and application of a self-referencing glucose microsensor for the measurement of glucose consumption by pancreatic beta-cells. Anal Chem 2001; 73:3759-67. [PMID: 11510845 DOI: 10.1021/ac010247u] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Glucose gradients generated by an artificial source and beta-cells were measured using an enzyme-based glucose microsensor, 8-microm tip diameter, as a self-referencing electrode. The technique is based on a difference measurement between two locations in a gradient and thus allows us to obtain real-time flux values with minimal impact of sensor drift or noise. Flux values were derived by incorporation of the measured differential current into Fick's first equation. In an artificial glucose gradient, a flux detection limit of 8.2 +/- 0.4 pmol.cm(-2).s(-1) (mean +/- SEM, n = 7) with a sensor sensitivity of 7.0 +/- 0.4 pA/ mM (mean +/- SEM, n = 16) was demonstrated. Under biological conditions, the glucose sensor showed no oxygen dependence with 5 mM glucose in the bulk medium. The addition of catalase to the bulk medium was shown to ameliorate surface-dependent flux distortion close to specimens, suggesting an underlying local accumulation of hydrogen peroxide. Glucose flux from beta-cell clusters, measured in the presence of 5 mM glucose, was 61.7 +/- 9.5 fmol.nL(-1).s(-1) (mean +/- SEM, n = 9) and could be pharmacologically modulated. Glucose consumption in response to FCCP (1 microM) transiently increased, subsequently decreasing to below basal by 93 +/- 16 and 56 +/- 6%, respectively (mean +/- SEM, n = 5). Consumption was decreased after the application of 10 microM rotenone by 74 +/- 5% (mean +/- SEM, n = 4). These results demonstrate that an enzyme-based amperometric microsensor can be applied in the self-referencing mode. Further, in obtaining glucose flux measurements from small clusters of cells, these are the first recordings of the real-time dynamic of glucose movements in a biological microenvironment.
Collapse
Affiliation(s)
- S K Jung
- Marine Biological Laboratory, BioCurrents Research Center, Woods Hole, Massachusetts 02543, USA
| | | | | | | |
Collapse
|