601
|
Weaver LC, Marsh DR, Gris D, Meakin SO, Dekaban GA. Central mechanisms for autonomic dysreflexia after spinal cord injury. PROGRESS IN BRAIN RESEARCH 2002; 137:83-95. [PMID: 12440361 DOI: 10.1016/s0079-6123(02)37009-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Lynne C Weaver
- Spinal Cord Injury Laboratory, BioTherapeutics Research Group, John P. Robarts Research Institute, 100 Perth Drive, P.O. Box 5015, London, ON N6A 5K8, Canada.
| | | | | | | | | |
Collapse
|
602
|
Lacroix S, Chang L, Rose-John S, Tuszynski MH. Delivery of hyper-interleukin-6 to the injured spinal cord increases neutrophil and macrophage infiltration and inhibits axonal growth. J Comp Neurol 2002; 454:213-28. [PMID: 12442313 DOI: 10.1002/cne.10407] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cytokine growth factors of the interleukin (IL)-6 family have recently been shown to play an important role in central nervous system (CNS) development, repair, and inflammation. These cytokines, which interact via specific membrane receptors, share a signal-transducing receptor subunit, glycoprotein 130 (gp130). Gp130 is expressed by motoneurons in the gray matter of the rat spinal cord and by several brainstem nuclei that project to the spinal cord including the red, reticular, and vestibular nuclei. In this study, we examined whether stimulation of gp130 signaling, with the use of grafts of fibroblasts genetically modified to deliver the fusion protein, hyper-IL-6 (H-IL-6), which consists of the cytokine growth factor, IL-6, and its alpha receptor, would elicit growth of injured spinal cord axons. Particular emphasis was placed on examining the potentially competing effects of growth factor versus proinflammatory influences of H-IL-6 in the context of spinal cord injury. Our results demonstrated that grafts delivering H-IL-6 induce a sixfold increase in the number of neutrophils (P < 0.05) and a twofold increase in the areas of spinal tissue occupied by macrophages and activated microglia (P < 0.01) at the site of the spinal cord injury when compared with control grafts. Of note, this augmentation in inflammatory cell infiltration correlated with a significant twofold increase in lesion size (P < 0.05) and a fourfold reduction in axonal growth (P < 0.01) at the lesion site. Thus, potential neurotrophic properties of this cytokine family of growth factors must be balanced against their inflammatory properties when considering therapeutic application to CNS injury.
Collapse
Affiliation(s)
- Steve Lacroix
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
603
|
Novikova LN, Novikov LN, Kellerth JO. Differential effects of neurotrophins on neuronal survival and axonal regeneration after spinal cord injury in adult rats. J Comp Neurol 2002; 452:255-63. [PMID: 12353221 DOI: 10.1002/cne.10381] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Spinal cord injury (SCI) induces retrograde cell death in descending pathways, which can be prevented by long-term intrathecal infusion of neurotrophins (Novikova et al. [2000] Eur J Neurosci 12:776-780). The present study investigates whether the same treatment also leads to improved regeneration of the injured tracts. After cervical SCI in adult rats, a peripheral nerve graft was attached to the rostral wall of the lesion cavity. The animals were treated by local application into the cavity of Gelfoam soaked in (1) phosphate buffered saline (untreated controls) or (2) a mixture of the neurotrophins brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) (local treatment), or by intrathecal infusion of BDNF + NT-3 for (3) 2 weeks (short-term treatment) or (4) 5-8 weeks (long-term treatment). Despite a very strong survival effect, long-term treatment failed to stimulate ingrowth of descending tracts into the nerve graft. In comparison with untreated controls, the latter treatment also caused 35% reduction in axonal sprouting of descending pathways rostral to the lesion site and 72% reduction in the number of spinal cord neurons extending axons into the nerve graft. Local and short-term treatments neither prevented retrograde cell death nor enhanced regeneration of descending tracts, but induced robust regeneration of spinal cord neurons into the nerve graft. These results indicate that the signal pathways promoting neuronal survival and axonal regeneration, respectively, in descending tracts after SCI respond differently to neurotrophic stimuli and that efficient rescue of axotomized tract neurons is not a sufficient prerequisite for regeneration.
Collapse
Affiliation(s)
- Liudmila N Novikova
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, SE-901 87 Umeå, Sweden
| | | | | |
Collapse
|
604
|
Earnhardt JN, Streit WJ, Anderson DK, O'Steen WA, Nick HS. Induction of manganese superoxide dismutase in acute spinal cord injury. J Neurotrauma 2002; 19:1065-79. [PMID: 12482119 DOI: 10.1089/089771502760341974] [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: 12/12/2022] Open
Abstract
Free radical-mediated mechanisms of cellular damage have been implicated in the early stages of spinal cord injury (SCI). Manganese superoxide dismutase (MnSOD) is a potent scavenger of superoxide radicals and likely serves an important cytoprotective role in preventing cellular damage after SCI. We have evaluated the expression of MnSOD to address its role during the early events of SCI using a well-established rat contusion model. Northern analysis showed a rapid induction of MnSOD mRNA between 2 and 6 h post injury. Observed time-dependent increases in MnSOD message was maximal 6 h post injury over that of MnSOD mRNA levels induced by laminectomy alone. Immunoblot and immunohistochemical analysis demonstrated increased expression of MnSOD protein 24 h after SCI with localization primarily within neurons. Interestingly, laminectomy alone also caused an induction of MnSOD gene and protein expression. To evaluate one potential mechanism of MnSOD induction, we microinjected the naive spinal cord with IL-1beta, which caused a similar fold induction of MnSOD mRNA levels by 6 h as observed with SCI, thus implicating it as a potential inducer of MnSOD during SCI. In summary, these results demonstrate that this potent cytoprotective antioxidant enzyme is rapidly and significantly induced as a consequence of SCI.
Collapse
Affiliation(s)
- J N Earnhardt
- Department of Neuroscience, University of Florida College of Medicine, Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, Florida 32610, USA
| | | | | | | | | |
Collapse
|
605
|
Penkowa M, Poulsen C, Carrasco J, Hidalgo J. M-CSF deficiency leads to reduced metallothioneins I and II expression and increased tissue damage in the brain stem after 6-aminonicotinamide treatment. Exp Neurol 2002; 176:308-21. [PMID: 12359172 DOI: 10.1006/exnr.2002.7968] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
6-Aminonicotinamide (6-AN) is a niacin antagonist, which leads to degeneration of gray-matter astrocytes followed by a vigorous inflammatory response. Macrophage colony stimulating factor (M-CSF) is important during inflammation, and in order to further clarify the roles for M-CSF in neurodegeneration and brain cell death, we have examined the effect of 6-AN on osteopetrotic mice with genetic M-CSF deficiency (op/op mice). The 6-AN-induced degeneration of gray-matter areas was comparable in control and op/op mice, but the numbers of reactive astrocytes, macrophages, and lymphocytes in the damaged areas were significantly decreased in op/op mice relative to controls. The levels of oxidative stress (as determined by using immunoreactivity for inducible nitric oxide synthase, nitrotyrosine, and malondialdehyde) and apoptotic cell death (as determined by using TUNEL and immunoreactivity for caspases and cytochrome c) were significantly increased in 6-AN-injected op/op mice relative to controls. From a number of antioxidant factors assayed, only metallothioneins I and II (MT-I+II) were decreased in op/op mice in comparison to controls. Thus, the present results indicate that M-CSF is an important growth factor for coping with 6-AN-induced central nervous system damage and suggest that MT-I+II are likely to have a significant role.
Collapse
Affiliation(s)
- Milena Penkowa
- Department of Medical Anatomy, The Panum Institute, University of Copenhagen, Denmark.
| | | | | | | |
Collapse
|
606
|
Popovich PG, Guan Z, McGaughy V, Fisher L, Hickey WF, Basso DM. The neuropathological and behavioral consequences of intraspinal microglial/macrophage activation. J Neuropathol Exp Neurol 2002; 61:623-33. [PMID: 12125741 DOI: 10.1093/jnen/61.7.623] [Citation(s) in RCA: 246] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Activated microglia and macrophages (CNS macrophages) have been implicated in the secondary or "bystander" pathology (e.g. axon injury, demyelination) that accompanies traumatic or autoimmune injury to the brain and spinal cord. These cells also can provide neurotrophic support and promote axonal regeneration. Studying the divergent functional potential of CNS macrophages in trauma models is especially difficult due to the various degradative mechanisms that are initiated prior to or concomitant with microglial/macrophage activation (e.g. hemorrhage, edema, excitotoxicity, lipid peroxidation). To study the potential impact of activated CNS macrophages on the spinal cord parenchyma, we have characterized an in vivo model of non-traumatic spinal cord neuroinflammation. Specifically, focal activation of CNS macrophages was achieved using stereotaxic microinjections of zymosan. Although microinjection does not cause direct mechanical trauma, localized activation of macrophages with zymosan acts as an "inflammatory scalpel" causing tissue injury at and nearby the injection site. The present data reveal that activation of CNS macrophages in vivo can result in permanent axonal injury and demyelination. Moreover, the pathology can be graded and localized to specific white matter tracts to produce quantifiable behavioral deficits. Further development of this model will help to clarify the biological potential of microglia and macrophages and the molecular signals that control their function within the spinal cord.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Antigens, Neoplasm
- Antigens, Surface
- Avian Proteins
- Axons/drug effects
- Axons/metabolism
- Axons/pathology
- Basigin
- Blood Proteins
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/physiology
- Denervation/methods
- Disease Models, Animal
- Female
- Gait Disorders, Neurologic/chemically induced
- Gait Disorders, Neurologic/pathology
- Gait Disorders, Neurologic/physiopathology
- Gliosis/chemically induced
- Gliosis/pathology
- Gliosis/physiopathology
- Immunohistochemistry
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/metabolism
- Membrane Glycoproteins/metabolism
- Microglia/cytology
- Microglia/drug effects
- Microglia/metabolism
- Microinjections
- Myelin Sheath/drug effects
- Myelin Sheath/metabolism
- Myelin Sheath/pathology
- Myelitis/chemically induced
- Myelitis/pathology
- Myelitis/physiopathology
- Nerve Degeneration/chemically induced
- Nerve Degeneration/pathology
- Nerve Degeneration/physiopathology
- Nerve Fibers, Myelinated/drug effects
- Nerve Fibers, Myelinated/pathology
- Rats
- Rats, Sprague-Dawley
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spinal Cord/physiopathology
- Spinal Cord Injuries/pathology
- Spinal Cord Injuries/physiopathology
- Zymosan/pharmacology
Collapse
Affiliation(s)
- P G Popovich
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University College of Medicine & Public Health and School of Allied Medical Professions, Columbus 43210, USA
| | | | | | | | | | | |
Collapse
|
607
|
Ma M, Wei T, Boring L, Charo IF, Ransohoff RM, Jakeman LB. Monocyte recruitment and myelin removal are delayed following spinal cord injury in mice with CCR2 chemokine receptor deletion. J Neurosci Res 2002; 68:691-702. [PMID: 12111830 DOI: 10.1002/jnr.10269] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The inflammatory response initiated after spinal cord injury (SCI) is characterized by the accumulation of macrophages at the impact site. Monocyte chemoattractant protein-1 (MCP-1) is a strong candidate for mediating chemotaxis of monocytes to the injured nervous system. To help in defining the role of MCP-1 in inflammation after SCI, we evaluated the time course of macrophage accumulation for 2 weeks following a midthoracic spinal cord contusion injury in mice lacking CCR2, a principal receptor for MCP-1. Mice with a deletion of CCR2 resulted in significantly reduced Mac-1 immunoreactivity restricted to the lesion epicenter at 7 days postinjury. The regions devoid of Mac-1 immunoreactivity corresponded to areas of reduced myelin degradation at this time. By 14 days postinjury, however, there were no differences in Mac-1 staining between CCR2 (+/+) and CCR2 (-/-) mice. Analyses of mRNA levels by RNase protection assay (RPA) revealed increases in MCP-1 as well as MCP-3 and MIP-2 mRNA at 1 day postinjury compared with 7 day postinjury. There were no differences in chemokine expression between CCR2-deficient mice and wild-type littermate controls. The CCR2-deficient mice also exhibited reduced expression of mRNA for chemokine receptors CCR1 and CCR5. Together, these results indicate that chemokines acting through CCR2 contribute to the early recruitment of monocytes to the lesion epicenter following SCI.
Collapse
Affiliation(s)
- Manhong Ma
- Department of Physiology and Cell Biology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|
608
|
Murakami Y, Furukawa S, Nitta A, Furukawa Y. Accumulation of nerve growth factor protein at both rostral and caudal stumps in the transected rat spinal cord. J Neurol Sci 2002; 198:63-9. [PMID: 12039665 DOI: 10.1016/s0022-510x(02)00080-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Changes in the nerve growth factor (NGF) content in the rat spinal cord during development or after traumatic spinal cord injury were examined by using a two-site enzyme immunoassay (EIA) system and an immunohistochemical technique. From embryonic day (E) 14 to postnatal day (P) 70, the spinal cord contained 200-300 pg NGF/g of wet tissue evenly in all regions tested. After complete spinal cord transection of P49 rats, the NGF level started to increase in the rostral and caudal stumps nearest to the injury site at 2 and 4 days, respectively. The NGF level of the caudal side returned to the original level by 2 weeks, but that of the rostral side remained high even 3 weeks, after the injury. At 4 days after the injury, NGF-like immunoreactivity in both stumps was predominantly localized in the axon-like structures of the white matter and in cells morphologically resembling immune cells. These observations suggest that the NGF was transported within the spinal tracts, and that NGF secreted from immune cells that had invaded into the injured spinal cord had accumulated around the transection site. Increased NGF at the injury site may be advantageous for injured neurons and involved in mechanisms directing to axonal regeneration of the injured spinal cord.
Collapse
Affiliation(s)
- Yutaka Murakami
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Mitahora-Higashi, Japan
| | | | | | | |
Collapse
|
609
|
Abstract
Both acute and chronic inflammatory processes have been shown to influence outcome in experimental models of spinal cord injury. Although early inflammatory responses may participate in secondary injury processes, more delayed inflammatory events may be reparative. Therapeutic strategies that target these events are currently based on experimental findings that have clarified the cellular and molecular processes involved in the inflammatory response to injury. An increasing body of literature supports the hypothesis that acute inflammatory events are attenuated by therapeutic hypothermia and other anti-inflammatory strategies, whereas immune neuroprotection and axonal regeneration can be achieved by transfer of activated T cells or by treatment with therapeutic vaccines. These data are summarized in the present review.
Collapse
Affiliation(s)
- John R Bethea
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Lois Pope LIFE Center, 1095 NW 14th Terrace-(R-48), Miami, FL 33136, USA.
| | | |
Collapse
|
610
|
Affiliation(s)
- Richard M Ransohoff
- The Lerner Research Institute, 9500 Euclid Avenue, Room NC30, Cleveland, OH 44195, USA.
| |
Collapse
|
611
|
Takami T, Oudega M, Bethea JR, Wood PM, Kleitman N, Bunge MB. Methylprednisolone and interleukin-10 reduce gray matter damage in the contused Fischer rat thoracic spinal cord but do not improve functional outcome. J Neurotrauma 2002; 19:653-66. [PMID: 12042099 DOI: 10.1089/089771502753754118] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The effects of two antiinflammatory and neuroprotective agents, methylprednisolone (MP) and interleukin-10 (IL-10), singly and in combination on tissue damage, axonal preservation and functional recovery were studied in the contused adult Fischer rat thoracic spinal cord 12 weeks after injury. MP (30 mg/kg at 5 min, and 2 and 4 h after injury) was administered intravenously and IL-10 (15 or 30 microg/kg at 30 min after injury), intraperitoneally. MP, IL-10, or the combination significantly reduced the volume of damaged tissue (including cavities) compared to control animals. The loss of spinal tissue (cavities) was reduced after treatment with MP alone or combined with IL-10, but not with IL-10 alone. The reduction in tissue damage was confined to spinal gray matter; at the level of the lesion epicenter, the thickness of the lateral white matter columns was similar in all groups. Retrograde tracing using fast blue revealed that the number of spared propriospinal and supraspinal projections was similar in all groups at 12 weeks after the contusion. The open-field BBB-test showed no significant difference in hindlimb locomotion between groups. Our results demonstrate that all tested antiinflammatory treatments significantly increase the volume of spared spinal gray matter 3 months after a moderate contusion of the Fischer rat thoracic spinal cord, but none of the treatments improved axonal preservation or functional recovery.
Collapse
Affiliation(s)
- Toshihiro Takami
- The Chambers Family Laboratory of Electron Microscopy, The Miami Project to Cure Paralysis, Miami, Florida, USA
| | | | | | | | | | | |
Collapse
|
612
|
Hostettler ME, Knapp PE, Carlson SL. Platelet-activating factor induces cell death in cultured astrocytes and oligodendrocytes: involvement of caspase-3. Glia 2002; 38:228-39. [PMID: 11968060 DOI: 10.1002/glia.10065] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The biologically active lipid metabolite, platelet-activating factor (PAF), is thought to contribute to inflammatory processes and tissue damage in a variety of central nervous system (CNS) injuries. In previous studies, we found that after contusion spinal cord injury, treatment with a PAF antagonist led to significantly increased white matter tissue sparing as well as decreased mRNA levels for pro-inflammatory cytokines. Some studies suggest that PAF can also have toxic effects on neurons in vitro. Few studies, however, have examined the effects of PAF on glial cells of the CNS. In the present study, the potential for PAF to act as a toxin to cultured astrocytes was examined. Also investigated were the effects of PAF on oligodendrocytes at two different stages of development. Treatment with 0.02-2 microM PAF for 72 h resulted in significant levels of cell death in both cell types (P < 0.05), an effect that was blocked by the PAF receptor antagonists, WEB 2170 and BN 52021. To investigate PAF-induced glial cell death further, we looked for activation of the enzyme, caspase-3, which can be indicative of apoptosis. Immunocytochemistry demonstrated that PAF at all concentrations caused activation of caspase-3 at 24, 48, and 72 h after treatment in both cell types. Caspase-3-dependent cell death was further confirmed using knockout mice (-/-) deficient in the caspase-3 gene. Toxicity was lost when astrocytes (-/-) were exposed to 0.02-2 microM PAF (P < 0.01). Oligodendrocytes (-/-) were not susceptible to toxicity at 2 microM PAF (P < 0.001). The results demonstrate that the pro-inflammatory molecule, PAF, induces cell death in cultured CNS glial cells and that this effect is, in part, dependent on caspase-3 activation.
Collapse
Affiliation(s)
- Mary Ellen Hostettler
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Lexington 40536-0398, USA
| | | | | |
Collapse
|
613
|
Sayer FT, Oudega M, Hagg T. Neurotrophins reduce degeneration of injured ascending sensory and corticospinal motor axons in adult rat spinal cord. Exp Neurol 2002; 175:282-96. [PMID: 12009779 DOI: 10.1006/exnr.2002.7901] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Spinal cord regeneration in adult mammals is limited by neurite outgrowth inhibitors and insufficient availability of outgrowth-promoting agents. Formation of degenerative swellings at the proximal ends of severed axons (terminal clubs), which starts early after injury, also may hinder recovery and their rupture may contribute to secondary spinal cord damage. We investigated whether neurotrophins would reduce these degenerative processes. Adult rats received a transection of the dorsal column sensory and corticospinal motor tracts at T9 and anterograde tracing of the axons from the sciatic nerve and motor cortex, respectively. The highest number of terminal clubs was found at 1 day and approximately half remained present until at least 28 days. A single injection immediately after injury of a mixture of nerve growth factor, brain-derived neurotrophic factor and neurotrophin-3 into the lesion site, reduced the number of terminal clubs in the sensory system by approximately half at 1 and 7 days (but not 14) after the lesion. Individual or combinations of two neurotrophins were as effective, suggesting that the neurotrophins protected similar axonal populations. The injected neurotrophins did not affect degeneration of corticospinal motor axons. A 7-day continuous intrathecal infusion of neurotrophin-3 was more effective and also reduced terminal club formation of corticospinal axons by approximately 60%. Spinal tissue loss was not affected by the neurotrophin treatments, suggesting that terminal clubs are not major contributors to the pathogenesis of secondary spinal degeneration during the first two weeks. Thus, neurotrophins can reduce axonal degeneration in the spinal cord after traumatic axonal injury.
Collapse
Affiliation(s)
- Faisal T Sayer
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Kentucky 40292, USA
| | | | | |
Collapse
|
614
|
Chatzipanteli K, Garcia R, Marcillo AE, Loor KE, Kraydieh S, Dietrich WD. Temporal and segmental distribution of constitutive and inducible nitric oxide synthases after traumatic spinal cord injury: effect of aminoguanidine treatment. J Neurotrauma 2002; 19:639-51. [PMID: 12042098 DOI: 10.1089/089771502753754109] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nitric oxide (NO) has been shown to play an important role in the pathophysiology of traumatic brain injury (TBI) and cerebral ischemia. However, its contribution to the pathogenesis of traumatic spinal cord injury (SCI) remains to be clarified. This study determined the time course of constitutive and inducible nitric oxide synthases (cNOS and iNOS, respectively) after SCI. Rats underwent moderate SCI at T10 using the NYU impactor device and were allowed to survive for 3, 6, or 24 h and 3 days after SCI (n = 5 in each group). For the determination of enzymatic activities, spinal cords were dissected into five segments, including levels rostral and caudal (remote) to the injury site. Other rats were perfusion fixed for the immunohistochemical localization of iNOS protein levels. cNOS activity was significantly decreased at 3 and 6 h within the traumatized T10 segment and at 3, 6, and 24 h at the rostral (T9) level (p < 0.05). Rostral (T8) and caudal (T11, T12) to the injury site cNOS activity was also decreased at 3 h after injury (p < 0.05). However, cNOS activity returned to control levels within 6 h at T8, T11 and T12 and at one day at T10 and T9 segments. iNOS enzymatic activity was elevated at all time points tested (p < 0.05), with the most robust increase observed at 24 h. Immunostaining for iNOS at 24 h revealed that a significant cellular source of iNOS protein appeared to be invading polymorphonuclear leukocytes (PMNLs). To assess the functional consequences of iNOS inhibition, aminoguanidine treatment was initiated 5 min after SCI and rats tested using the BBB open field locomotor score. Treated rats demonstrated significantly improved hindlimb function up to 7 weeks after SCI. Histopathological analysis of contusion volume showed that aminoguanidine treatment decreased lesion volume by 37% (p < 0.05). In conclusion, these results indicate that (1) cNOS and iNOS activities are regionally and temporally affected after moderate SCI, (2) the early accumulation of PMNLs are a potentially significant source of NO-induced cytotoxic products, and (3) acute aminoguanidine treatment significantly improves functional and histopathological outcome after SCI.
Collapse
Affiliation(s)
- Katina Chatzipanteli
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, Florida 33101, USA.
| | | | | | | | | | | |
Collapse
|
615
|
Pathological CNS autoimmune disease triggered by traumatic spinal cord injury: implications for autoimmune vaccine therapy. J Neurosci 2002. [PMID: 11923434 DOI: 10.1523/jneurosci.22-07-02690.2002] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Lymphocytes respond to myelin proteins after spinal cord injury (SCI) and may contribute to post-traumatic secondary degeneration. However, there is increasing evidence that autoreactive T-lymphocytes may also convey neuroprotection and promote functional recovery after CNS injury. To clarify the role of myelin autoreactive lymphocytes after SCI, we performed contusion injuries in the thoracic spinal cord of transgenic (Tg) mice in which >95% of all CD4+ T-lymphocytes are reactive with myelin basic protein (MBP). We observed significantly impaired recovery of locomotor and reflex function in Tg mice compared with non-Tg (nTg) littermates. Measures of functional impairment in Tg mice correlated with significantly less white matter at the injury site, and morphometric comparisons of injured Tg and nTg spinal cords revealed increased rostrocaudal lesion expansion (i.e., secondary degeneration) in Tg mice. Rostrocaudal to the impact site in SCI-nTg mice, demyelination was restricted to the dorsal funiculus, i.e., axons undergoing Wallerian degeneration. The remaining white matter appeared normal. In contrast, lymphocytes were colocalized with regions of demyelination and axon loss throughout the white matter of SCI-Tg mice. Impaired neurological function and exacerbated neuropathology in SCI-Tg mice were associated with increased intraspinal production of proinflammatory cytokine mRNA; neurotrophin mRNA was not elevated. These data suggest that endogenous MBP-reactive lymphocytes, activated by traumatic SCI, can contribute to tissue injury and impair functional recovery. Any neuroprotection afforded by myelin-reactive T-cells is likely to be an indirect effect mediated by other non-CNS-reactive lymphocytes. Similar to the Tg mice in this study, a subset of humans that are genetically predisposed to autoimmune diseases of the CNS may be adversely affected by vaccine therapies designed to boost autoreactive lymphocyte responses after CNS trauma. Consequently, the safe implementation of such therapies requires that future studies define the mechanisms that control T-cell function within the injured CNS.
Collapse
|
616
|
Abstract
BACKGROUND CONTEXT Recent advances in neuroscience have opened the door for hope toward prevention and cure of the devastating effects of spinal cord injury (SCI). PURPOSE To highlight the current understanding of traumatic SCI mechanisms, provide information regarding state-of-the-art care for the acute spinal cord-injured patient, and explore future treatments aimed at neural preservation and reconstruction. STUDY DESIGN/SETTING A selective overview of the literature pertaining to the neuropathophysiology of traumatic SCI is provided with an emphasis on pharmacotherapies and posttraumatic experimental strategies aimed at improved neuropreservation and late neuroregenerative repair. METHODS One hundred fifty-four peer-reviewed basic science and clinical articles pertaining to SCI were reviewed. Articles cited were chosen based on the relative merits and contribution to the current understanding of SCI neuropathophysiology, neuroregeneration, and clinical SCI treatment patterns. RESULTS A better understanding of the pathophysiology and early treatment for the spinal cord-injured patient has led to a continued decrease in mortality, decreased acute hospitalization and complication rates, and more rapid rehabilitation and re-entry into society. Progressive neural injury results from a combination of secondary injury mechanisms, including ischemia, biochemical alterations, apoptosis, excitotoxicity, calpain proteases, neurotransmitter accumulation, lipid peroxidation/free radical injury, and inflammatory responses. Experimental studies suggest that the final posttraumatic neurologic deficit is not only a result of the initial impaction forces but rather a combination of these forces and secondary time-dependent events that follow shortly after the initial impact. CONCLUSIONS Experimental studies continue to provide a better understanding of the complex interaction of pathophysiologic events after traumatic SCI. Future approaches will involve strategies aimed at blocking the multiple mechanisms of progressive central nervous system injury and promoting neuroregeneration.
Collapse
Affiliation(s)
- Gregory D Carlson
- Department of Orthopaedic Surgery, Reeve-Irvine Research Center, University California, Irvine, Long Beach Veterans Administration, 5901 East 7th Street, Long Beach, CA 90822, USA.
| | | |
Collapse
|
617
|
Liao B, Newmark H, Zhou R. Neuroprotective effects of ginseng total saponin and ginsenosides Rb1 and Rg1 on spinal cord neurons in vitro. Exp Neurol 2002; 173:224-34. [PMID: 11822886 DOI: 10.1006/exnr.2001.7841] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Spinal cord injury is a major cause of disability and results in many serious physical, psychological, and social difficulties. Numerous studies have shown that traumatic spinal cord injuries (SCI) lead to neuronal loss and axonal degeneration in and around the injury site that cause partial disability or complete paralysis. An important strategy in the treatment of SCI is to promote neuron survival and axon outgrowth, making possible the recovery of neural connections. Using an in vitro survival assay, we have identified ginsenosides Rb1 and Rg1, extracted from ginseng root (Panax ginseng C. A. Meyer), as efficient neuroprotective agents for spinal cord neurons. These compounds protect spinal neurons from excitotoxicity induced by glutamate and kainic acid, as well as oxidative stress induced by H(2)O(2). The neuroprotective effects are dose-dependent. The optimal doses are 20-40 microM for ginsenosides Rb1 and Rg1. The effects are specific for Rb1 and Rg1, since a third ginsenoside, Re, did not exhibit any activity. Ginseng has been used for thousands of years in the treatment of neurological disorders and other diseases in Asia. Ginsenosides Rb1 and Rg1 represent potentially effective therapeutic agents for spinal cord injuries.
Collapse
Affiliation(s)
- Baisong Liao
- Laboratory for Cancer Research, Department of Chemical Biology, College of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
618
|
David S, Ousman SS. Recruiting the immune response to promote axon regeneration in the injured spinal cord. Neuroscientist 2002; 8:33-41. [PMID: 11843097 DOI: 10.1177/107385840200800108] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Myelin contains molecules that can inhibit the growth and regeneration of axons. Neutralizing the activity of these inhibitors can enhance axon regeneration in the adult mammalian central nervous system (CNS). The complexity of the CNS-immune system interactions after CNS trauma is now beginning to be better understood. Recent studies indicate that both cell-mediated and antibody-mediated immune responses can help in promoting axon regeneration after CNS injury. It is hoped that such advances will lead to the development of safe and effective vaccine and cytokine treatments for spinal cord injuries.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, Montreal General Hospital Research Institute, McGill University, Quebec, Canada.
| | | |
Collapse
|
619
|
Kojima A, Tator CH. Intrathecal administration of epidermal growth factor and fibroblast growth factor 2 promotes ependymal proliferation and functional recovery after spinal cord injury in adult rats. J Neurotrauma 2002; 19:223-38. [PMID: 11893024 DOI: 10.1089/08977150252806974] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have shown previously that epidermal growth factor (EGF) plus fibroblast growth factor (FGF2) expands the neural precursor cells in the ependyma of the normal adult rat spinal cord in vivo. To investigate the therapeutic effect of these factors on spinal cord injury (SCI), we administered EGF, FGF2, EGF plus FGF2, or artificial cerebrospinal fluid (aCSF) intrathecally (15 ng/h of EGF or FGF2) for 3 or 14 days after mild (2.4-g) or moderate (20-g) clip compression injury at T1 in adult rats. Histological and functional assessments were used to evaluate the therapeutic effects. The EGF plus FGF2 group, which received these agents for 14 days, showed better functional recovery than the aCSF group 42 days after moderate SCI (p < 0.05). At 14 days, the EGF plus FGF2 group showed a much greater expansion of ependymal cells and astrocytes compared to the other groups, and there was evidence for extensive migration of ependymal cells into the surrounding injured cord. These mitogens did not significantly enhance nestin expression in the ependymal layer or alter the expansion of oligodendrocyte precursor cells or microglia/macrophages, and dividing cells did not show the neuron-specific marker NeuN except immediately adjacent to the ependyma. The exact mechanism for improved functional recovery after EGF plus FGF2 is not known.
Collapse
Affiliation(s)
- Atsuhiro Kojima
- Division of Neurosurgery, University of Toronto and Toronto Western Institute, Ontario, Canada
| | | |
Collapse
|
620
|
Tzeng SF, Bresnahan JC, Beattie MS, de Vellis J. Upregulation of the HLH Id gene family in neural progenitors and glial cells of the rat spinal cord following contusion injury. J Neurosci Res 2001; 66:1161-72. [PMID: 11746449 DOI: 10.1002/jnr.10089] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Spinal cord injury (SCI) leads to a complex sequence of cellular responses, including astrocyte activation, oligodendrocyte death, and ependymal cell proliferation. Inhibitors of DNA binding (Id1, Id2, Id3) belong to a helix-loop-helix (HLH) gene family. Id genes have been implicated in playing a vital role in the proliferation of many cell types, including astrocytes and myoblasts. In the present study, the expression of Id family members in spinal cord after contusion injury was investigated by in situ hybridization. Id1, Id2, and Id3 mRNA expression was upregulated 5 mm rostral and caudal to the lesion center, and reached maximal levels 3 days after SCI. In addition, cell populations expressing Id1, Id2, and Id3 mRNA were maximally increased 3 days after SCI. The increase in Id2 and Id3 mRNA expression and Id2 and Id3 mRNA+ cells was still observed at 8 days. The Id mRNA expressing cells were phenotyped by combining immunostaining of cell-specific markers with in situ hybridization. Glial fibrillary acidic protein (GFAP)+ astrocytes were found to express all three Id mRNA, whereas S-100alpha+ astrocytes only expressed high levels of Id2 and Id3 mRNA. Cells having a neural progenitor morphology and the marker nestin appeared after SCI and they expressed Id1, Id2, and Id3 mRNA. Interestingly, some Rip+ oligodendrocytes located in the areas close to the central canal expressed Id3 mRNA after injury. In conclusion, Id genes are upregulated in a time-dependent manner in astrocytes, oligodendrocytes, and neural progenitor subpopulations after SCI, suggesting that they play major roles in cellular responses following SCI.
Collapse
Affiliation(s)
- S F Tzeng
- Department of Neurobiology, Mental Retardation Research Center, UCLA School of Medicine, 760 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | | | | | | |
Collapse
|
621
|
Yamamoto S, Yamamoto N, Kitamura T, Nakamura K, Nakafuku M. Proliferation of parenchymal neural progenitors in response to injury in the adult rat spinal cord. Exp Neurol 2001; 172:115-27. [PMID: 11681845 DOI: 10.1006/exnr.2001.7798] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has long been believed that the fully developed mammalian central nervous system (CNS) lacks significant regenerative capacity. Recent advances have revealed, however, that many regions of the adult CNS contain neural progenitors that have the ability to generate new neurons and glia. Although the periventricular area has been identified as a rich source of these progenitors, their precise location in each region and details of their properties in vivo still remain poorly understood. Here we provide evidence that in the adult rat spinal cord, a significant number of neural progenitors are present, not only in the periventricular area, but also in other regions of the parenchyma. These progenitors could proliferate in vitro as neurosphere-like cell aggregates in the presence of growth factors and also gave rise to neurons and glia under appropriate conditions. We further demonstrate that these parenchymal neural progenitors were capable of proliferating in vivo in response to injury. Immunohistochemical studies suggested that proliferative progenitors emerged throughout the gray and white matter in the lesioned spinal cord. Consistently, an increased number of neurosphere-forming cells could be isolated from injured tissues, and they were able to differentiate into neurons in vitro. The widespread occurrence of neural progenitors in the parenchyma expands the possibility of repairing damaged tissue by activating the latent regenerative potential of the adult spinal cord.
Collapse
Affiliation(s)
- S Yamamoto
- Department of Neurobiology, University of Tokyo Graduate School of Medicine, Tokyo 113-0033, Japan
| | | | | | | | | |
Collapse
|
622
|
Hutchinson KJ, Linderman JK, Basso DM. Skeletal muscle adaptations following spinal cord contusion injury in rat and the relationship to locomotor function: a time course study. J Neurotrauma 2001; 18:1075-89. [PMID: 11686494 DOI: 10.1089/08977150152693764] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Experimental spinal cord injury (SCI) via contusion of moderate severity results in residual locomotor deficits, including a lack of coordination and trunk stability. Given that muscle contractile properties and fiber composition adapt to reduced neural input and/or weight bearing, contusion-induced locomotor deficits may reflect changes in hindlimb skeletal muscle. Therefore, we examined muscle adaptations during early (1 week), intermediate (3 week), and late (10 week) stages of motor recovery after moderate SCI. Forty-two Sprague Dawley rats underwent SCI via 1.1mm cord displacement with the OSU impact device or served as age and weight-matched or laminectomy controls. Subsets of rats had soleus (SOL) in vitro physiological testing or SOL and extensor digitorum longus (EDL) myosin heavy chain (MHC) fiber type analysis. At 1 week post-SCI during paralysis/paresis, a significant decrease in wet weight occurred in the plantaris, medial/lateral gastrocnemius (MG/LG), tibialis anterior, and SOL. Changes in contractile properties of the SOL did not accompany muscle wet weight changes. By 3 weeks, the loss of weight-bearing activity early after SCI induced significant decreases in SOL peak twitch and peak tetanic tension as well as significantly greater IIx MHC expression in the EDL. By 10 weeks post-SCI, after several weeks of weight supported stepping, muscle wet weight, contractile properties and MHC composition returned to baseline levels except for MG/LG atrophy. Thus, muscle plasticity appears to be extremely sensitive to locomotor deficits and their resolution after moderate spinal cord contusion.
Collapse
Affiliation(s)
- K J Hutchinson
- Physical Therapy Department, Northeastern University, Boston, Massachusetts, USA
| | | | | |
Collapse
|
623
|
Kyrkanides S, O'Banion MK, Whiteley PE, Daeschner JC, Olschowka JA. Enhanced glial activation and expression of specific CNS inflammation-related molecules in aged versus young rats following cortical stab injury. J Neuroimmunol 2001; 119:269-77. [PMID: 11585630 DOI: 10.1016/s0165-5728(01)00404-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Aging is associated with increased glial responsiveness that may enhance the brain's susceptibility to injury and disease. To determine whether unique age-related molecular responses occur in brain injury, we assessed mRNA levels of representative central nervous system (CNS) inflammation-related molecules in young (3 months) and aged (36 months) Fisher 344/Brown Norwegian F1 hybrid rats following cortical stab. Enhanced glial activation in older animals was accompanied by increased expression of a subset of inflammation-related mRNAs, including IL-1beta, TNFalpha, IL-6, ICAM-1, inducible nitric oxide synthase (iNOS), metalloproteinase-9 (MMP-9), and complement 3alpha-chain 1 (C3alpha1). Recognition of these age-specific differences may guide development of novel treatment regimes for older individuals.
Collapse
Affiliation(s)
- S Kyrkanides
- Department of Neurobiology and Anatomy, University of Rochester, 601 Elmwood Avenue, Box 603, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
624
|
Schwab JM, Frei E, Klusman I, Schnell L, Schwab ME, Schluesener HJ. AIF-1 expression defines a proliferating and alert microglial/macrophage phenotype following spinal cord injury in rats. J Neuroimmunol 2001; 119:214-22. [PMID: 11585624 DOI: 10.1016/s0165-5728(01)00375-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Microglial cells are among the first and dominant cell types to respond to CNS injury. Following calcium influx, microglial activation leads to a variety of cellular responses, such as proliferation and release of cytotoxic and neurotrophic mediators. Allograft inflammatory factor-1, AIF-1 is a highly conserved EF-handed, putative calcium binding peptide, associated with microglia activation in the brain. Here, we have analyzed the expression of AIF-1 following spinal cord injury at the lesion site and at remote brain regions. Following spinal cord injury, AIF-1+ cells accumulated in parenchymal pan-necrotic areas and perivascular Virchow-Robin spaces. Subsequent to culmination at day 3--a situation characterized by infiltrating blood borne macrophages and microglia activation--AIF-1+ cell numbers decreased until day 7. In remote areas of Wallerian degeneration and delayed neuronal death, a more discrete and delayed activation pattern of AIF-1+ microglia/macrophages reaching maximum levels at day 14 was observed. There was a considerable match between AIF-1+ cells and PCNA (proliferating cell nuclear antigen) or Ki-67+ labeled cells. AIF-1 expression preceded the expression of ED1, thus indicating a pre-phagocytic role. It appears that AIF-1+ microglia/macrophages are among the earliest cells to respond to spinal cord injury. Our results suggest a role of AIF-1 in the initiation of the early microglial response leading to activation and proliferation essential for the acute response to CNS injury. AIF-1 might modulate microgliosis influencing the efficacy of tissue debris removal, myelin degradation, recruitment of oligodendrocytes and re-organisation of the CNS architecture.
Collapse
Affiliation(s)
- J M Schwab
- Institute of Brain Research, University of Tuebingen, Medical School, Calwer Str. 3, D-72076, Tuebingen, Germany.
| | | | | | | | | | | |
Collapse
|
625
|
Hill CE, Beattie MS, Bresnahan JC. Degeneration and sprouting of identified descending supraspinal axons after contusive spinal cord injury in the rat. Exp Neurol 2001; 171:153-69. [PMID: 11520130 DOI: 10.1006/exnr.2001.7734] [Citation(s) in RCA: 217] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Contusive spinal cord injury (SCI) results in the formation of a chronic lesion cavity surrounded by a rim of spared fibers. Tissue bridges containing axons extend from the spared rim into the cavity dividing it into chambers. Whether descending axons can grow into these trabeculae or whether fibers within the trabeculae are spared fibers remains unclear. The purposes of the present study were (1) to describe the initial axonal response to contusion injury in an identified axonal population, (2) to determine whether and when sprouts grow in the face of the expanding contusion cavity, and (3) in the long term, to see whether any of these sprouts might contribute to the axonal bundles that have been seen within the chronic contusion lesion cavity. The design of the experiment also allowed us to further characterize the development of the lesion cavity after injury. The corticospinal tract (CST) underwent extensive dieback after contusive SCI, with retraction bulbs present from 1 day to 8 months postinjury. CST sprouting occurred between 3 weeks and 3 months, with penetration of CST axons into the lesion matrix occurring over an even longer time course. Collateralization and penetration of reticulospinal fibers were observed at 3 months and were more extensive at later time points. This suggests that these two descending systems show a delayed regenerative response and do extend axons into the lesion cavity and that the endogenous repair can continue for a very long time after SCI.
Collapse
Affiliation(s)
- C E Hill
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
626
|
Schwab JM, Seid K, Schluesener HJ. Traumatic brain injury induces prolonged accumulation of cyclooxygenase-1 expressing microglia/brain macrophages in rats. J Neurotrauma 2001; 18:881-90. [PMID: 11565600 DOI: 10.1089/089771501750451802] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inflammatory cellular responses to brain injury are promoted by proinflammatory messengers. Cyclooxygenases (prostaglandin endoperoxide H synthases [PGH]) are key enzymes in the conversion of arachidonic acid into prostanoids, which mediate immunomodulation, mitogenesis, apoptosis, blood flow, secondary injury (lipid peroxygenation), and inflammation. Here, we report COX-1 expression following brain injury. In control brains, COX-1 expression was localized rarely to brain microglia/macrophages. One to 5 days after injury, we observed a highly significant (p < 0.0001) increase in COX-1+ microglia/macrophages at perilesional areas and in the developing core with a delayed culmination of cell accumulation at day 7, correlating with phagocytic activity. There, cell numbers remained persistently elevated up to 21 days following injury. Further, COX-1+ cells were located in perivascular Virchow-Robin spaces also reaching maximal numbers at day 7. Lesion-confined COX-1+ vessels increased in numbers from day 1, reaching the maximum at days 5-7. Double-labeling experiments confirmed coexpression of COX-1 by ED-1+ and OX-42+ microglia/ macrophages. Transiently after injury, most COX-1+ microglia/macrophages coexpress the activation antigen OX-6 (MHC class II). However, the prolonged accumulation of COX-1+, ED-1+ microglia/macrophages in lesional areas enduring the acute postinjury inflammatory response points to a role of COX-1 in the pathophysiology of secondary injury. We have identified localized, accumulated COX-1 expression as a potential pharmacological target in the treatment of brain injury. Our results suggest that therapeutic approaches based on long-term blocking including COX-1, might be superior to selective COX-2 blocking to suppress the local synthesis of prostanoids.
Collapse
Affiliation(s)
- J M Schwab
- Institute of Brain Research, University of Tuebingen, Medical School, Germany.
| | | | | |
Collapse
|
627
|
Popovich PG, Stuckman S, Gienapp IE, Whitacre CC. Alterations in immune cell phenotype and function after experimental spinal cord injury. J Neurotrauma 2001; 18:957-66. [PMID: 11565606 DOI: 10.1089/089771501750451866] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Traumatic injury to the spinal cord initiates a cascade of inflammatory-mediated injury and repair processes within the nervous system. In parallel, spinal injury could influence peripheral mechanisms of host defense (e.g., wound healing, antibody production) by altering lymphocyte phenotype and function. The goal of this study was to evaluate the physiological impact of spinal contusion injury on phenotypic and functional indices of lymphocyte activation. A flow cytometric time-course analysis of lymphocytes isolated from lymph node and spleen revealed an increase in CD4+ and a decrease in CD8+ lymphocytes during the first week post injury. The functional potential of lymphocytes was also evaluated based on their ability to proliferate in the presence of a biologically relevant antigen (myelin basic protein, MBP) or a lymphocyte mitogen. The data revealed increased proliferation to MBP by 3 days postinjury in lymphocytes isolated from lymph node but not spleen. By 1 week postinjury, increased proliferation to mitogen was noted in both the lymph node and the spleen suggesting a general increase in lymphocyte reactivity during this time interval. Circulating corticosterone (CORT), an endogenous glucocorticoid with significant effects on lymphocyte phenotype and function, was elevated within 24 h after spinal cord injury (SCI) and remained above control levels throughout the duration of our studies (up to 1 month postinjury). The present data suggest injury-associated changes in immune cell phenotype and function paralleled by the activation of the hypothalamic-pituitary-adrenal (HPA) axis.
Collapse
Affiliation(s)
- P G Popovich
- Department of Molecular Virology, Immunology & Medical Genetics, Ohio State University College of Medicine and Public Health, Columbus, 43210, USA.
| | | | | | | |
Collapse
|
628
|
Tsai EC, van Bendegem RL, Hwang SW, Tator CH. A novel method for simultaneous anterograde and retrograde labeling of spinal cord motor tracts in the same animal. J Histochem Cytochem 2001; 49:1111-22. [PMID: 11511680 DOI: 10.1177/002215540104900905] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Examination of repaired spinal cord tracts has usually required separate groups of animals for anterograde and retrograde tracing owing to the incompatibility of techniques such as tissue fixation. However, anterograde and retrograde labeling of different animals subjected to the same repair may not allow accurate examination of that repair strategy because widely variable results can occur in animals subjected to the same strategy. We have developed a reliable method of labeling spinal cord motor tracts bidirectionally in the same animal using DiI, a lipophilic dye, to anterogradely label the corticospinal tract and Fluoro-Gold (FG) to retrogradely label cortical and brainstem neurons of several spinal cord motor tracts in normal and injured adult rats. Other tracer combinations (lipophilic dyes or fluorescent dextrans) were also investigated but were less effective. We also developed methods to minimize autofluorescence with the DiI/FG technique, and found that the DiI/FG technique is compatible with decalcification and immunohistochemistry for several markers relevant for studies of spinal cord regeneration. Thus, the use of anterograde DiI and retrograde FG is a novel technique for bidirectional labeling of the motor tracts of the adult spinal cord with fluorescent tracers and should be useful for demonstrating neurite regeneration in studies of spinal cord repair.(J Histochem Cytochem 49:1111-1122, 2001)
Collapse
Affiliation(s)
- E C Tsai
- Toronto Western Hospital Research Institute and University of Toronto, 399 Bathurst Street, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
629
|
Abstract
Support is provided for the hypothesis that activated leukocytes, especially monocytes/macrophages, contribute to cerebral white matter damage in extremely low gestational age newborns. Much of the evidence is indirect and comes from analogies to brain diseases in adults, and from models of brain damage in adult and newborn animals. If the recruitment of circulating cells to the brain contributes to white matter damage in extremely low gestational age newborns, then minimizing the transendothelial migration of circulating cells by pharmacological manipulation might prevent or reduce the occurrence of neonatal white matter damage and the disabilities that follow.
Collapse
Affiliation(s)
- O Dammann
- Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
630
|
Lenzlinger PM, Morganti-Kossmann MC, Laurer HL, McIntosh TK. The duality of the inflammatory response to traumatic brain injury. Mol Neurobiol 2001; 24:169-81. [PMID: 11831551 DOI: 10.1385/mn:24:1-3:169] [Citation(s) in RCA: 311] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
One and a half to two million people sustain a traumatic brain injury (TBI) in the US each year, of which approx 70,000-90,000 will suffer from long-term disability with dramatic impacts on their own and their families' lives and enormous socio-economic costs. Brain damage following traumatic injury is a result of direct (immediate mechanical disruption of brain tissue, or primary injury) and indirect (secondary or delayed) mechanisms. These secondary mechanisms involve the initiation of an acute inflammatory response, including breakdown of the blood-brain barrier (BBB), edema formation and swelling, infiltration of peripheral blood cells and activation of resident immunocompetent cells, as well as the intrathecal release of numerous immune mediators such as interleukins and chemotactic factors. An overview over the inflammatory response to trauma as observed in clinical and in experimental TBI is presented in this review. The possibly harmful/beneficial sequelae of post-traumatic inflammation in the central nervous system (CNS) are discussed using three model mediators of inflammation in the brain, tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and transforming growth factor-beta (TGF-beta). While the former two may act as important mediators for the initiation and the support of post-traumatic inflammation, thus causing additional cell death and neurologic dysfunction, they may also pave the way for reparative processes. TGF-beta, on the other hand, is a potent anti-inflammatory agent, which may also have some deleterious long-term effects in the injured brain. The implications of this duality of the post-traumatic inflammatory response for the treatment of brain-injured patients using anti-inflammatory strategies are discussed.
Collapse
Affiliation(s)
- P M Lenzlinger
- Department of Neurosurgery, University of Pennsylvania, Veterans Administration Medical Center, Philadelphia 19104-6316, USA
| | | | | | | |
Collapse
|
631
|
Abstract
Pain is classically viewed as being mediated solely by neurons, as are other sensory phenomena. The discovery that spinal cord glia (microglia and astrocytes) amplify pain requires a change in this view. These glia express characteristics in common with immune cells in that they respond to viruses and bacteria, releasing proinflammatory cytokines, which create pathological pain. These spinal cord glia also become activated by certain sensory signals arriving from the periphery. Similar to spinal infection, these signals cause release of proinflammatory cytokines, thus creating pathological pain. Taken together, these findings suggest a new, dramatically different approach to pain control, as all clinical therapies are focused exclusively on altering neuronal, rather than glial, function.
Collapse
Affiliation(s)
- L R Watkins
- Dept of Psychology & the Center for Neurosciences, University of Colorado at, Boulder, CO, USA.
| | | | | |
Collapse
|
632
|
MIP-1alpha, MCP-1, GM-CSF, and TNF-alpha control the immune cell response that mediates rapid phagocytosis of myelin from the adult mouse spinal cord. J Neurosci 2001. [PMID: 11425892 DOI: 10.1523/jneurosci.21-13-04649.2001] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The slow immune response in the adult mammalian CNS results in slow myelin phagocytosis along degenerating white matter after injury. This has important consequences for axon regeneration because of the presence of axon growth inhibitors in myelin. In addition, abnormal immune cell responses in the CNS lead to demyelinating disease. Lysophosphatidylcholine (LPC) can induce an inflammatory response in the CNS, producing rapid demyelination without much damage to adjacent cells. In this study, we searched for the molecular switches that turn on this immune cell response. Using reverse transcription PCR analysis, we show that mRNA expression of macrophage inflammatory protein-1alpha (MIP-1alpha), macrophage chemotactic protein-1 (MCP-1), granulocyte macrophage-colony stimulating factor (GM-CSF), and tumor necrosis factor-alpha (TNF-alpha) in the spinal cord is rapidly and transiently upregulated after intraspinal injection of LPC. Neutralizing these signaling molecules with function-blocking antibodies suppresses recruitment of T-cells, neutrophils, and monocytes into the spinal cord, as well as significantly reduces the number of phagocytic macrophages and the demyelination induced by LPC. These findings will have important implications for CNS regeneration and demyelinating disease.
Collapse
|
633
|
Ma M, Basso DM, Walters P, Stokes BT, Jakeman LB. Behavioral and histological outcomes following graded spinal cord contusion injury in the C57Bl/6 mouse. Exp Neurol 2001; 169:239-54. [PMID: 11358439 DOI: 10.1006/exnr.2001.7679] [Citation(s) in RCA: 189] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A computer-controlled electromagnetic spinal cord injury device (ESCID) has been adapted to develop a mouse model of spinal cord contusion injury. In the present study, we have extended this model in C57Bl/6 mice with behavioral and histopathological outcome assessment. Three groups of mice received a laminectomy at the T(9) vertebral level followed by a contusion injury from a predetermined starting load of 1500 dynes. Contusion was produced by rapid displacement of the spinal cord to a peak distance of 0.3, 0.5, or 0.8 mm, with the entire injury and retraction procedure completed over a 23-ms epoch. Control groups received laminectomy alone or complete transection. Functional recovery was examined for 9 weeks after injury using the BBB locomotor rating scale, grid walking, and footprint analysis. Distinct patterns of locomotor recovery were evident across the five groups. Measurements of spared white matter at the epicenter, lesion length, and cross-sectional area of fibronectin-immunopositive scar tissue were also significantly different between injury groups. The severity of injury corresponded with the biomechanical measures recorded at the time of impact as well as with behavioral and histological parameters. The results demonstrate that graded contusion injuries can be produced reliably in mice using the ESCID. The data provide a thorough and quantitative analysis of the effects of contusion injury on long-term behavioral and histological outcome measures in this strain and species.
Collapse
Affiliation(s)
- M Ma
- Department of Physiology and Cell Biology, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
634
|
Popovich PG, Hickey WF. Bone marrow chimeric rats reveal the unique distribution of resident and recruited macrophages in the contused rat spinal cord. J Neuropathol Exp Neurol 2001; 60:676-85. [PMID: 11444796 DOI: 10.1093/jnen/60.7.676] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Brain and spinal cord inflammation that develops after traumatic injury is believed to differentially influence the structural and/or physiological integrity of surviving neurons and glia. It is possible that the functional dichotomy of CNS inflammation results from the activity of a heterogeneous macrophage population elicited by trauma. Indeed, unique functions have been attributed to macrophages derived from resident microglia versus those originating from infiltrating monocytes. Thus, whether progressive tissue injury or repair is favored could be explained by the disproportionate contributions of one macrophage subset relative to the other. Descriptive neuroanatomical studies are a reasonable first approach to revealing a relationship between microglia, recruited blood monocytes/macrophages, and regions of tissue degeneration and/or repair. Unfortunately, it is not possible to differentiate between CNS macrophage subsets using conventional immunohistochemical approaches. In the present study, we have used radiation bone marrow chimeric rats to definitively characterize the macrophage reaction elicited by experimental spinal contusion injury. In chimeric animals, antibodies raised against unique cell surface molecules expressed on bone marrow-derived cells (BMCs) were used to distinguish infiltrating BMCs from resident microglial-derived macrophages. Our findings indicate that the onset and plateau of macrophage activation (previously shown to be 3 and 7 days postinjury, respectively) is dominated initially by microglial-derived macrophages and then is supplanted by hematogenous cells. While resident macrophages are ubiquitously distributed throughout the injury site, leukocyte-derived monocytes exclusively infiltrate the gray matter and to a lesser extent subpial white matter. Generally, monocyte foci in white matter remain associated with the lumen or abluminal surface of blood vessels, i.e. few cells actually infiltrate the parenchyma. If functional differences exist between CNS macrophage subsets, differences in the time-dependent accumulation and distribution of these cell types could differentially influence the survival of surrounding neurons and glia.
Collapse
Affiliation(s)
- P G Popovich
- Department of Molecular Virology, Immunology & Medica Genetics, The Ohio State University College of Medicine and Public Health, Columbus, USA
| | | |
Collapse
|
635
|
Abstract
Primary damage caused by injury to the CNS is often followed by delayed degeneration of initially spared neurons. Studies in our laboratory have shown that active or passive immunization with CNS myelin-associated self-antigens can reduce this secondary loss. Here we show, using four experimental paradigms in rodents, that CNS trauma spontaneously evokes a beneficial T cell-dependent immune response, which reduces neuronal loss. (1) Survival of retinal ganglion cells in rats was significantly higher when optic nerve injury was preceded by an unrelated CNS (spinal cord) injury. (2) Locomotor activity of rat hindlimbs (measured in an open field using a locomotor rating scale) after contusive injury of the spinal cord (T8) was significantly better (by three to four score grades) after passive transfer of myelin basic protein (MBP)-activated splenocytes derived from spinally injured rats than in untreated injured control rats or rats similarly treated with splenocytes from naive animals or with splenocytes from spinally injured rats activated ex vivo with ovalbumin or without any ex vivo activation. (3) Neuronal survival after optic nerve injury was 40% lower in adult rats devoid of mature T cells (caused by thymectomy at birth) than in normal rats. (4) Retinal ganglion cell survival after optic nerve injury was higher (119 +/- 3.7%) in transgenic mice overexpressing a T cell receptor (TcR) for MBP and lower (85 +/- 1.3%) in mice overexpressing a T cell receptor for the non-self antigen ovalbumin than in matched wild types. Taken together, the results imply that CNS injury evokes a T cell-dependent neuroprotective response.
Collapse
|
636
|
Abstract
Given the numerous reparative roles glia may play after spinal cord injury (SCI), glial proliferation and cell number were examined in a model of traumatic SCI. Emphasis was placed on analysis of oligodendrocytes and NG2-positive (NG2+) cells, an endogenous cell population that may be involved in oligodendrocyte replacement. Overall, proliferation (assessed by bromodeoxyuridine incorporation) was markedly elevated during the first 2 weeks after injury and declined thereafter; a large portion of these dividing cells likely consisted of microglia-macrophages. Although the total number of NG2+ cells in the epicenter was reduced by half, we noted protracted proliferation in surviving NG2+ cells, with values sevenfold greater than in uninjured controls. Elevated proliferation of NG2+ cells persisted throughout the first 4 weeks after injury. However, the absolute number of NG2+ cells was not increased over controls, suggesting that the daughter cells either did not survive or they differentiated into other cell types. As expected, oligodendrocyte numbers were drastically altered after SCI. By 7 d after injury, the number of oligodendrocytes at the impact site was reduced by 93%. Despite ongoing tissue loss, the number of oligodendrocytes in spared tissue rose threefold at 14 d after injury. Although the function of NG2+ cells within the spinal cord is not completely understood, several studies suggest that they may differentiate into oligodendrocytes. Thus, proliferating NG2+ cells may contribute to the increased oligodendrocyte number observed at 2 weeks after injury. Future studies are required, however, to definitively determine the role NG2+ cells play in oligodendrocyte genesis, remyelination, and other post-injury events.
Collapse
|
637
|
Schwartz M, Kipnis J. Protective autoimmunity: regulation and prospects for vaccination after brain and spinal cord injuries. Trends Mol Med 2001; 7:252-8. [PMID: 11378514 DOI: 10.1016/s1471-4914(01)01993-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Neuronal degeneration after traumatic injury to the central nervous system (CNS) can be reduced by active immunization or passive transfer of T cells against CNS-associated myelin antigens. We propose that a protective autoimmunity is evoked by CNS insult when non-immunological local protective mechanisms cannot adequately buffer the injury-induced toxicity. The ability of a particular strain to develop a protective autoimmune response appears to be inversely related to its susceptibility to autoimmune disease. We also propose that vaccination with specific CNS-derived'safe' (non-pathogenic) peptides after traumatic CNS insult, and possibly at any stage of chronic neurodegenerative disease, can be used to boost the protective autoimmunity and thereby to reduce further injury-induced damage. Such therapeutic vaccination ensures that the augmented beneficial autoimmunity will be free of accompanying disease.
Collapse
Affiliation(s)
- M Schwartz
- Department of Neurobiology, The Weizmann Institute of Science, 76100, Rehovot, Israel.
| | | |
Collapse
|
638
|
Casha S, Yu WR, Fehlings MG. Oligodendroglial apoptosis occurs along degenerating axons and is associated with FAS and p75 expression following spinal cord injury in the rat. Neuroscience 2001; 103:203-18. [PMID: 11311801 DOI: 10.1016/s0306-4522(00)00538-8] [Citation(s) in RCA: 293] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Apoptosis or programmed cell death has been reported after CNS trauma. However, the significance of this mechanism in the pathophysiology of spinal cord injury, in particular at the cervical level, requires further investigation. In the present study, we used the extradural clip compression model in the rat to examine the cellular distribution of apoptosis following cervical spinal cord injury, the relationship between glial apoptosis and post-traumatic axonal degeneration and the possible role of apo[apoptosis]-1, CD95 (FAS) and p75 in initiating post-traumatic glial apoptosis. In situ terminal-deoxy-transferase mediated dUTP nick end labeling revealed apoptotic cells, largely oligodendrocytes as identified by cell specific markers, in grey and white matter following spinal cord injury. Apoptotic cell death was confirmed using electron microscopy and by the demonstration of DNA laddering on agarose gel electrophoresis. Beta-amyloid precursor protein was used as a molecular marker of axonal degeneration on western blots and immunohistochemistry. Degeneration of axons was temporally and spatially co-localized with glial apoptosis. FAS and p75 protein expression was seen in astrocytes, oligodendrocytes and microglia, and was also seen in some apoptotic glia after cord injury. Both FAS and p75 increased in expression in a temporal course, which mirrored the development of cellular apoptosis. The downstream caspases 3 and 8, which are linked to FAS and p75, demonstrated activation at times of maximal apoptosis, while FLIP-L an inhibitor of caspase 8, decreased at times of maximal apoptosis. We conclude that axonal degeneration after traumatic spinal cord injury is associated with glial, in particular oligodendroglial, apoptosis. Activation of the FAS and p75 death receptor pathways may be involved in initiating this process.
Collapse
Affiliation(s)
- S Casha
- Division of Neurosurgery, Toronto Western Research, The University Health Network, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
639
|
Xu J, Kim GM, Chen S, Yan P, Ahmed SH, Ku G, Beckman JS, Xu XM, Hsu CY. iNOS and nitrotyrosine expression after spinal cord injury. J Neurotrauma 2001; 18:523-32. [PMID: 11393255 DOI: 10.1089/089771501300227323] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Secondary tissue damage after spinal cord injury (SCI) may be due to inflammatory mediators. After SCI, the nuclear factor-kappaB (NF-kappaB) transcription factor can activate many pro-inflammatory genes, one of which is inducible nitric oxide synthase (iNOS). iNOS catalyzes the synthesis of nitric oxide (NO), a key inflammatory mediator, which in turn reacts with superoxide to generate peroxynitrite. Peroxynitrite is a strong oxidant that can damage cellular enzymes, membranes, and subcellular organelles through the nitration of tyrosine residues on proteins. The presence of nitrotyrosine (NT) is an indirect chemical indicator of toxic NO and peroxynitrite-induced cellular damage. Using a New York University (NYU) impactor to induce SCI in adult rats, we examined the temporal and cellular expression of iNOS and NT. We observed a progressive increase in iNOS expression in the injured cord starting at day 1 with maximal expression occurring at day 7, as determined by Western blot analysis. iNOS expression corresponded temporally to an increase in iNOS enzyme activity after SCI. In parallel with the progressive increase in iNOS activity, NT expression also increased with time after SCI. The iNOS and NT immunoreactivity was localized in neurons, astrocytes, endothelial cells and ependymal cells at the epicenter and adjacent to the region of spinal cord impact and injury. Results from the present study suggest that increased iNOS and peroxynitrite anion, as reflected by the progressive accumulation of NT in the injured impacted spinal cord, may contribute to the secondary injury process after SCI.
Collapse
Affiliation(s)
- J Xu
- Department of Neurology and Center for the Study of Nervous System Injury, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
640
|
Lenzlinger PM, Hans VH, Jöller-Jemelka HI, Trentz O, Morganti-Kossmann MC, Kossmann T. Markers for cell-mediated immune response are elevated in cerebrospinal fluid and serum after severe traumatic brain injury in humans. J Neurotrauma 2001; 18:479-89. [PMID: 11393251 DOI: 10.1089/089771501300227288] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The brain is believed to be an immunologically privileged organ, sheltered from the systemic immunological defense by the blood-brain barrier (BBB). However, there is increasing evidence for a marked inflammatory response in the brain after traumatic brain injury (TBI). Markers for cellular immune activation, neopterin, beta2-microglobulin (beta2M), and soluble interleukin-2 receptor (sIL-2R), were measured for up to 3 weeks in cerebrospinal fluid (CSF) and serum of 41 patients with severe TBI in order to elucidate the time course and the origin of the cellular immune response following TBI. Neopterin gradually increased during the first posttraumatic week in both CSF and serum. Concentrations in CSF were generally higher than in serum, suggesting intrathecal release of this marker. beta2M showed similar kinetics but with higher serum than CSF concentrations. Nonetheless, intrathecal release as assessed by the beta2M index could be postulated for most of the patients. The mean levels of sIL-2R in both CSF and serum were elevated during the whole study period, serum concentrations being up to 2 x 10(4) times higher than in CSF. No significant intrathecal production of sIL-2R could be detected. The present data shows that severe TBI leads to a marked cell-mediated immune response within the brain and in the systemic circulation. In the intrathecal compartment the activated cells appear to be predominantly of the macrophage/microglia lineage, while the immune activation in the systemic circulation seems to involve mainly T-lymphocytes.
Collapse
Affiliation(s)
- P M Lenzlinger
- Division of Trauma Surgery, University Hospital, Zurich, Switzerland.
| | | | | | | | | | | |
Collapse
|
641
|
Abstract
Injuries of the central nervous system (CNS) lead to an inevitable and irreversible loss of function because of the lack of neurogenesis, poor regeneration, and the spread of degeneration. In most tissues, protection and repair are the function of the immune system. It has long been thought that this does not apply to the CNS, where--because of its immune-privileged character--any immune activity was assumed to be detrimental. We have recently proposed, however, that provided care is taken to avoid the attendant risks, both repair and protection of injured CNS neurons can benefit from immune intervention. In the following I will summarize the data that led to this concept and describe the evidence supporting it.
Collapse
Affiliation(s)
- M Schwartz
- Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| |
Collapse
|
642
|
Grossman SD, Rosenberg LJ, Wrathall JR. Relationship of altered glutamate receptor subunit mRNA expression to acute cell loss after spinal cord contusion. Exp Neurol 2001; 168:283-9. [PMID: 11259116 DOI: 10.1006/exnr.2001.7629] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alterations in the expression of ionotropic glutamate receptors (GluR) contribute to neuronal loss after brain ischemia and epilepsy. In order to determine whether altered expression of GluR subunits might contribute to cell loss after spinal cord injury (SCI), we performed a time course study of subunit mRNA expression using quantitative in situ hybridization. Expression was studied in ventral horn motor neurons (VMN) and glia in adjacent ventral white matter at 15 min and 4, 8, and 24 h after SCI in tissue sections 4 mm rostral and caudal to the injury epicenter. We found that the AMPA subunit GluR2 was significantly down-regulated in VMN at 24 h, but not at the earlier times examined, although half the loss of VMN in these locations occurs by 8 h after injury. No changes in the normal expression of GluR2 or GluR4 were found in white matter where glial loss occurs after SCI. NMDA subunits NR1 and NR2A were significantly and rapidly up-regulated in VMN after SCI, but only caudal to the lesion site, while VMN loss is similar rostral and caudal to the epicenter. Thus, the temporal pattern of AMPA and the spatial pattern of NMDA subunit expression changes were distinct from the pattern of VMN loss after SCI. We conclude that altered GluR subunit expression after SCI is unlikely to be involved in secondary cell loss and instead may be involved with plasticity and reorganization of the injured spinal cord.
Collapse
Affiliation(s)
- S D Grossman
- Department of Cell Biology, Georgetown University Medical Center, 3970 Reservoir Road, Washington, DC 20007, USA
| | | | | |
Collapse
|
643
|
Lu J, Moochhala S, Kaur C, Ling EA. Cellular inflammatory response associated with breakdown of the blood-brain barrier after closed head injury in rats. J Neurotrauma 2001; 18:399-408. [PMID: 11336441 DOI: 10.1089/089771501750170976] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study reports a widespread microglial response characterized by an upregulation of surface antigens, such as complement type 3 receptors (CR3) and major histocompatibility complex (MHC) class II antigens on these cells following closed head injury. Increased expression of CR3 (OX-42) and MHC class II antigens (OX-6) was observed in rats killed at 1, 3, and 5 days after injury. Intense OX-42 immunoreactivity was observed in microglial cells throughout the brain with a smaller number of them being OX-6 positive. In addition to microglial reaction, astrocytic activation reflected in cellular hypertrophy and increased immunoreactivity for glial fibrillary acidic protein (GFAP) was observed at 5 days after head injury. Together with the above, a diffuse perivascular and intraneuronal immunostaining for immunoglobulin G (IgG) was observed primarily in the cerebral cortex. This was accompanied by an enhanced expression of both endothelial nitric oxide synthase (eNOS) in blood vessels and inducible nitric oxide synthase (iNOS) in brain macrophages. In rats subjected to closed head injury followed by a single intraperitoneal (i.p.) injection of rhodamine isothiocyanate (RhIc), seepage of the fluorescent dye into the neuropil was observed. This had resulted in the labelling of the cortical neurons clearly demonstrating a breakdown of the blood-brain barrier (BBB). In the latter, it is conceivable that the ensuing leakage of plasma immunoglobulins and other serum-derived materials could induce the expression of MHC class II antigens on microglia. The mechanism causing the BBB dysfunction is not clear, although present results suggest that excessive release of nitric oxide (NO) may be a contributory factor. The widespread activation of microglia in rats after head injury suggests their involvement in increased endocytosis and immunological responses.
Collapse
Affiliation(s)
- J Lu
- Defence Science & Technology Agency, Defence Medical Research Institute, Singapore
| | | | | | | |
Collapse
|
644
|
Plunkett JA, Yu CG, Easton JM, Bethea JR, Yezierski RP. Effects of interleukin-10 (IL-10) on pain behavior and gene expression following excitotoxic spinal cord injury in the rat. Exp Neurol 2001; 168:144-54. [PMID: 11170729 DOI: 10.1006/exnr.2000.7604] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intraspinal injection of quisqualic acid (QUIS) produces excitotoxic injury with pathophysiological characteristics similar to those associated with ischemic and traumatic spinal cord injury (SCI). Responses to QUIS-induced injury include an inflammatory component, as well as the development of spontaneous and evoked pain behaviors. We hypothesized that QUIS-induced inflammation and subsequent gene expression contribute to the development and progression of pain-related behaviors and that blockade of inflammation-related gene expression leads to the amelioration of these behaviors. Using the QUIS model of spinal cord injury, we examined whether interleukin-10 (IL-10), a potent anti-inflammatory cytokine, is able to reduce mRNA levels of inflammatory and cell death-related genes leading to a reduction of pain behaviors. The results demonstrate that animals receiving systemic injection of IL-10, 30 minutes following QUIS-induced SCI, showed a significant delay in the onset of excessive grooming behavior, a significant reduction in grooming severity, and a significant reduction in the longitudinal extent of a pattern of neuronal loss within the spinal cord characterized as "grooming-type damage." QUIS injections also resulted in an increase in mRNA levels of interleukin-1 beta (IL-1 beta), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), CD95 ligand (CD95-L, also called FAS-L/APO-1L), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Results of QUIS injury plus IL-10 treatment resulted in a significant downregulation of IL1-beta and iNOS mRNA and these results were supported by Western blot analysis of protein levels following IL-10 treatment. These data suggest that IL-10 reduces inflammation and that targeting injury-induced inflammation is an effective strategy for limiting the extent of neuronal damage following excitotoxic SCI and thus the onset and progression of injury-induced pain behaviors.
Collapse
Affiliation(s)
- J A Plunkett
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | |
Collapse
|
645
|
McTigue DM, Popovich PG, Jakeman LB, Stokes BT. Strategies for spinal cord injury repair. PROGRESS IN BRAIN RESEARCH 2001; 128:3-8. [PMID: 11105664 DOI: 10.1016/s0079-6123(00)28002-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- D M McTigue
- Department of Physiology and Cell Biology, Ohio State University, Columbus 43210, USA
| | | | | | | |
Collapse
|
646
|
Bethea JR. Spinal cord injury-induced inflammation: a dual-edged sword. PROGRESS IN BRAIN RESEARCH 2001; 128:33-42. [PMID: 11105667 DOI: 10.1016/s0079-6123(00)28005-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- J R Bethea
- Miami Project to Cure Paralysis, University of Miami School of Medicine, FL 33136, USA.
| |
Collapse
|
647
|
Popovich PG. Immunological regulation of neuronal degeneration and regeneration in the injured spinal cord. PROGRESS IN BRAIN RESEARCH 2001; 128:43-58. [PMID: 11105668 DOI: 10.1016/s0079-6123(00)28006-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- P G Popovich
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine and Public Health, Ohio State University, Columbus 43210, USA.
| |
Collapse
|
648
|
Beattie MS, Li Q, Bresnahan JC. Cell death and plasticity after experimental spinal cord injury. PROGRESS IN BRAIN RESEARCH 2001; 128:9-21. [PMID: 11105665 DOI: 10.1016/s0079-6123(00)28003-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- M S Beattie
- Department of Neuroscience, Ohio State University, Columbus 43210, USA.
| | | | | |
Collapse
|
649
|
Butovsky O, Hauben E, Schwartz M. Morphological aspects of spinal cord autoimmune neuroprotection: colocalization of T cells with B7‐2 (
CD86
) and prevention of cyst formation. FASEB J 2001. [DOI: 10.1096/fsb2fj000550fje] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Oleg Butovsky
- Department of Neurobiology The Weizmann Institute of Science Rehovot Israel
| | - Ehud Hauben
- Department of Neurobiology The Weizmann Institute of Science Rehovot Israel
| | - Michal Schwartz
- Department of Neurobiology The Weizmann Institute of Science Rehovot Israel
| |
Collapse
|
650
|
Glucocorticoid receptor-mediated suppression of activator protein-1 activation and matrix metalloproteinase expression after spinal cord injury. J Neurosci 2001. [PMID: 11150324 DOI: 10.1523/jneurosci.21-01-00092.2001] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Post-traumatic inflammatory reaction may contribute to progressive tissue damage after spinal cord injury (SCI). Two key transcription factors, nuclear factor kappaB (NF-kappaB) and activator protein-1 (AP-1), are activated in inflammation. An increase in NF-kappaB binding activity has been shown in the injured spinal cord. We report activation of AP-1 after SCI. Electrophoretic mobility shift assay showed that AP-1 binding activity increased after SCI, starting at 1 hr, peaking at 8 hr, and declining to basal levels by 7 d. Methylprednisolone (MP) is the only therapeutic agent approved by the Food and Drug Administration for treating patients with acute traumatic SCI. MP reduced post-traumatic AP-1 activation. RU486, a glucocorticoid receptor (GR) antagonist, reversed MP inhibition of AP-1 activation. Immunostaining showed an increase in the expression of the Fos-B and c-Jun components of AP-1 in the injured cord. A c-fos antisense oligodeoxynucleotide (ODN) inhibited AP-1, but not NF-kappaB, activation after SCI. AP-1 and NF-kappaB can transactivate genes encoding matrix metalloproteinase-1 (MMP-1) and MMP-9. Western blotting and immunostaining show increased expression of MMP-1 and MMP-9 in the injured cord. MP inhibited MMP-1 and MMP-9 expression after SCI. RU486 reversed this MP effect. The c-fos antisense ODN, however, failed to suppress MMP-1 or MMP-9 expression. These findings demonstrate that MP may suppress post-traumatic inflammatory reaction by inhibiting both the AP-1 and NF-kappaB transcription cascades via a GR mechanism. Expression of inflammatory genes such as MMP-1 and MMP-9 that are transactivated jointly by AP-1 and NF-kappaB may not be suppressed by inhibiting only AP-1 activity.
Collapse
|