Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Neurosci ; 21(4): 1283-91, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160399

ABSTRACT

Developing motoneurons require trophic support from their target, the skeletal muscle. Despite a large number of neurotrophic molecules with survival-promoting activity for isolated embryonic motoneurons, those factors that are required for motoneuron survival during development are still not known. Cytokines of the ciliary neurotrophic factor (CNTF)-leukemia inhibitory factor (LIF) family have been shown to play a role in motoneuron (MN) survival. Importantly, in mice lacking the LIFRbeta or the CNTFRalpha there is a significant loss of MNs during embryonic development. Because genetic deletion of either (or both) CNTF or LIF fails, by contrast, to perturb MN survival before birth, it was concluded that another ligand exists that is functionally inactivated in the receptor deleted mice, resulting in MN loss during development. One possible candidate for this ligand is the CNTF-LIF family member cardiotrophin-1 (CT-1). CT-1 is highly expressed in embryonic skeletal muscle, secreted by myotubes, and promotes the survival of cultured embryonic mouse and rat MNs. Here we show that ct-1 deficiency causes increased motoneuron cell death in spinal cord and brainstem nuclei of mice during a period between embryonic day 14 and the first postnatal week. Interestingly, no further loss was detectable during the subsequent postnatal period, and nerve lesion in young adult ct-1-deficient mice did not result in significant additional loss of motoneurons, as had been previously observed in mice lacking both CNTF and LIF. CT-1 is the first bona fide muscle-derived neurotrophic factor to be identified that is required for the survival of subgroups of developing motoneurons.


Subject(s)
Cytokines/metabolism , Interleukin-6 , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neurodegenerative Diseases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Axotomy , Brain Stem/embryology , Brain Stem/metabolism , Brain Stem/pathology , Cell Death , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130 , Cytokines/deficiency , Cytokines/genetics , Cytokines/pharmacology , Dose-Response Relationship, Drug , Facial Nerve , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Leukemia Inhibitory Factor , Lymphokines/genetics , Lymphokines/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Neurons/drug effects , Motor Neurons/pathology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , RNA, Messenger/biosynthesis , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord/pathology
3.
J Neurosci ; 20(16): 6117-24, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934261

ABSTRACT

Spinal motoneurons (MNs) in the chick embryo undergo programmed cell death coincident with the establishment of nerve-muscle connections and the onset of synaptic transmission at the neuromuscular junction. Chronic treatment of embryos during this period with nicotinic acetylcholine receptor (nAChR)-blocking agents [e.g., curare or alpha-bungarotoxin (alpha-BTX)] prevents the death of MNs. Although this rescue effect has been attributed previously to a peripheral site of action of the nAChR-blocking agents at the neuromuscular junction (NMJ), because nAChRs are expressed in both muscle and spinal cord, it has been suggested that the rescue effect may, in fact, be mediated by a direct central action of nAChR antagonists. By using a variety of different nAChR-blocking agents that target specific muscle or neuronal nAChR subunits, we find that only those agents that act on muscle-type receptors block neuromuscular activity and rescue MNs. However, paralytic, muscular dysgenic mutant chick embryos also exhibit significant increases in MN survival that can be further enhanced by treatment with curare or alpha-BTX, suggesting that muscle paralysis may not be the sole factor involved in MN survival. Taken together, the data presented here support the argument that, in vivo, nAChR antagonists promote the survival of spinal MNs primarily by acting peripherally at the NMJ to inhibit synaptic transmission and reduce or block muscle activity. Although a central action of these agents involving direct perturbations of MN activity may also play a contributory role, further studies are needed to determine more precisely the relative roles of central versus peripheral sites of action in MN rescue.


Subject(s)
Apoptosis/drug effects , Motor Neurons/drug effects , Muscle, Skeletal/embryology , Neuromuscular Junction/embryology , Nicotinic Antagonists/pharmacology , Spinal Cord/embryology , Animals , Apoptosis/physiology , Bungarotoxins/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Chick Embryo , Curare/pharmacology , Motor Neurons/cytology , Motor Neurons/metabolism , Movement/drug effects , Movement/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Peripheral Nerves/drug effects , Peripheral Nerves/embryology , Peripheral Nerves/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure
4.
J Neurosci ; 20(13): 5001-11, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10864958

ABSTRACT

Because of discrepancies in previous reports regarding the role of glial cell line-derived neurotrophic factor (GDNF) in motoneuron (MN) development and survival, we have reexamined MNs in GDNF-deficient mice and in mice exposed to increased GDNF after in utero treatment or in transgenic animals overexpressing GDNF under the control of the muscle-specific promoter myogenin (myo-GDNF). With the exception of oculomotor and abducens MNs, the survival of all other populations of spinal and cranial MNs were reduced in GDNF-deficient embryos and increased in myo-GDNF and in utero treated animals. By contrast, the survival of spinal sensory neurons in the dorsal root ganglion and spinal interneurons were not affected by any of the perturbations of GDNF availability. In wild-type control embryos, all brachial and lumbar MNs appear to express the GDNF receptors c-ret and GFRalpha1 and the MN markers ChAT, islet-1, and islet-2, whereas only a small subset express GFRalpha2. GDNF-dependent MNs that are lost in GDNF-deficient animals express ret/GFRalpha1/islet-1, whereas many surviving GDNF-independent MNs express ret/GFRalpha1/GFRalpha2 and islet-1/islet-2. This indicates that many GDNF-independent MNs are characterized by the presence of GFRalpha2/islet-2. It seems likely that the GDNF-independent population represent MNs that require other GDNF family members (neurturin, persephin, artemin) for their survival. GDNF-dependent and -independent MNs may reflect subtypes with distinct synaptic targets and afferent inputs.


Subject(s)
Apoptosis/physiology , Brain/embryology , Drosophila Proteins , Motor Neurons/physiology , Nerve Growth Factors , Nerve Tissue Proteins/physiology , Spinal Cord/embryology , Animals , Brain/cytology , Cell Survival/drug effects , Crosses, Genetic , Embryonic and Fetal Development , Gestational Age , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/drug effects , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/physiology , Spinal Cord/cytology
5.
J Comp Neurol ; 412(3): 506-14, 1999 Sep 27.
Article in English | MEDLINE | ID: mdl-10441236

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a member of the serine protease inhibitor (serpin) superfamily that has been shown previously to promote the survival and/or differentiation of rat cerebellar granule neurons and human retinoblastoma cells in vitro. However, in contrast to most serpins, PEDF has no inhibitory activity against any known proteases, and its described biological activities do not appear to require the serpin-reactive loop located toward the carboxy end of the polypeptide. Because another serpin, protease nexin-1, has been shown to promote the in vivo survival and growth of motor neurons, the authors investigated the potential neurotrophic effects of PEDF on spinal cord motor neurons in highly enriched cultures and in vivo after injury. Here, it is shown that native bovine and recombinant human PEDF promoted the survival and differentiation (neurite outgrowth) of embryonic chick spinal cord motor neurons in vitro in a dose-dependent manner. A truncated form of PEDF that lacks approximately 62% of the carboxy end of the polypeptide comprising the homologous serpin-reactive loop also exhibited neurotrophic activities similar to those of the full-length protein. Furthermore, the data here showed that PEDF was transported retrogradely and prevented the death and atrophy of spinal motor neurons in the developing neonatal mouse after axotomy. These results indicate that PEDF exerts trophic effects on motor neurons, and, together with previous reports, these findings suggest that this protein may be useful as a pharmacologic agent to promote the development and maintenance of motor neurons. J. Comp. Neurol. 412:506-514, 1999. Published 1999 Wiley-Liss, Inc.


Subject(s)
Eye Proteins , Motor Neurons/drug effects , Nerve Growth Factors/pharmacology , Proteins/pharmacology , Serpins/pharmacology , Spinal Cord/drug effects , Animals , Animals, Newborn , Axotomy , Biological Transport , Cattle , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Humans , Mice , Motor Neurons/cytology , Neurites/drug effects , Recombinant Proteins/pharmacology , Spinal Cord/embryology , Spinal Cord/growth & development
6.
J Neuropathol Exp Neurol ; 58(5): 499-504, 1999 May.
Article in English | MEDLINE | ID: mdl-10331438

ABSTRACT

Several studies have shown that both neuronal and glial cells express functional thrombin receptors as well as prothrombin transcripts. Recently, we (and others) have shown that alpha-thrombin induces apoptotic cell death in different neuronal cell types, including motoneurons, in culture. Thrombin-induced effects on different cells are mediated through the cell surface protease-activated thrombin receptor, PAR-1. Furthermore, it has been shown that, in contrast to thrombin, which induces proteolysis of other proteins besides its receptor, the thrombin receptor agonist peptide, serine-phenylalanine-leucine-leucine-arginine-asparagine-proline (SFLLRNP), is only known to activate this receptor. However, whether activation of the thrombin receptor in vivo affects the development of spinal cord motoneurons is not known. Here, we show that treatment with a synthetic SFLLRNP peptide induced a dose-dependent degeneration and death of spinal motoneurons both in highly enriched cultures and in the developing chick embryo in vivo. However, cotreatment with caspase inhibitors completely abolished SFLLRNP-induced motoneuron death both in vitro and in vivo. These results suggest that developing motoneurons express functionally active PAR-1 whose activation leads to cell death through stimulation of the caspase family of proteins. Our findings also suggest a novel and deleterious role for PAR-like receptors in the central nervous system, different from their previously known functions in the vascular and circulatory system.


Subject(s)
Motor Neurons/enzymology , Motor Neurons/pathology , Nerve Degeneration/enzymology , Receptors, Thrombin/metabolism , Animals , Apoptosis/physiology , Cell Nucleus/pathology , Cell Survival/physiology , Chick Embryo , Motor Neurons/chemistry , Nerve Degeneration/pathology , Receptor, PAR-1 , Serine Endopeptidases/metabolism
7.
J Neurobiol ; 38(4): 571-80, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10084690

ABSTRACT

Previous reports have shown that neuronal and glial cells express functionally active thrombin receptors. The thrombin receptor (PAR-1), a member of a growing family of protease activated receptors (PARs), requires cleavage of the extracellular amino-terminus domain by thrombin to induce signal transduction. Studies from our laboratory have shown that PAR-1 activation following the addition of thrombin or a synthetic thrombin receptor activating peptide (TRAP) induces motoneuron cell death both in vitro and in vivo. In addition to increasing motoneuron cell death, PAR- 1 activation leads to decreases in the mean neurite length and side branching in highly enriched motoneuron cultures. It has been suggested that motoneuron survival depends on access to sufficient target-derived neurotrophic factors through axonal branching and synaptic contacts. However, whether the thrombininduced effects on motoneurons can be prevented by neurotrophic factors is still unknown. Using highly enriched avian motoneuron cultures, we show here that alone, soluble chick skeletal muscle extracts (CMX), brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and glial cell line-derived neurotrophic factor (GDNF) significantly increased motoneuron survival compared to controls, whereas nerve growth factor (NGF) did not have a significant effect on motoneuron survival. Furthermore, cotreatment with muscle-derived agents (i.e., CMX, BDNF, GDNF) significantly prevented the death of motoneurons induced by alpha-thrombin. Yet, non-muscle-derived agents (CNTF and NGF) had little or no significant effect in reversing thrombin-induced motoneuron death. CMX and CNTF significantly increased the mean length of neurites, whereas NGF, BDNF, and GDNF failed to enhance neurite outgrowth compared to controls. Furthermore, CMX and CNTF significantly prevented thrombin-induced inhibition of neurite outgrowth, whereas BDNF and GDNF only partially reversed thrombin-induced inhibition of neurite outgrowth. These findings show differential effects of neurotrophic factors on thrombin-induced motoneuron degeneration and suggest specific overlaps between the trophic and stress pathways activated by some neurotrophic agents and thrombin, respectively.


Subject(s)
Motor Neurons/cytology , Nerve Growth Factors/pharmacology , Thrombin/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Culture Techniques/methods , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor , Humans , Kinetics , Motor Neurons/drug effects , Motor Neurons/physiology , Nerve Tissue Proteins/pharmacology , Receptor, PAR-1 , Receptors, Thrombin/physiology , Recombinant Proteins/pharmacology , Signal Transduction
8.
J Neurobiol ; 36(3): 379-94, 1998 Sep 05.
Article in English | MEDLINE | ID: mdl-9733073

ABSTRACT

During development of the avian neuromuscular system, lumbar spinal motoneurons (MNs) innervate their muscle targets in the hindlimb coincident with the onset and progression of MN programmed cell death (PCD). Paralysis (activity blockade) of embryos during this period rescues large numbers of MNs from PCD. Because activity blockade also results in enhanced axonal branching and increased numbers of neuromuscular synapses, it has been postulated that following activity blockade, increased numbers of MNs can gain access to muscle-derived trophic agents that prevent PCD. An assumption of the access hypothesis of MN PCD is the presence of an activity-dependent, muscle-derived sprouting or branching agent. Several previous studies of sprouting in the rodent neuromuscular system indicate that insulin-like growth factors (IGFs) are candidates for such a sprouting factor. Accordingly, in the present study we have begun to test whether the IGFs may play a similar role in the developing avian neuromuscular system. Evidence in support of this idea includes the following: (a) IGFs promote MN survival in vivo but not in vitro; (b) neutralizing antibodies against IGFs reduce MN survival in vivo; (c) both in vitro and in vivo, IGFs increase neurite growth, branching, and synapse formation; (d) activity blockade increases the expression of IGF-1 and IGF-2 mRNA in skeletal muscles in vivo; (e) in vivo treatment of paralyzed embryos with IGF binding proteins (IGF-BPs) that interfere with the actions of endogenous IGFs reduce MN survival, axon branching, and synapse formation; (f) treatment of control embryos in vivo with IGF-BPs also reduces synapse formation; and (g) treatment with IGF-1 prior to the major period of cell death (i.e., on embryonic day 6) increases subsequent synapse formation and MN survival and potentiates the survival-promoting actions of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) administered during the subsequent 4- to 5-day period of PCD. Collectively, these data provide new evidence consistent with the role of the IGFs as activity-dependent, muscle-derived agents that play a role in regulating MN survival in the avian embryo.


Subject(s)
Apoptosis/physiology , Motor Neurons/physiology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Somatomedins/physiology , Animals , Cell Count , Cells, Cultured , Chick Embryo , Hindlimb/innervation , Immunohistochemistry , Insulin-Like Growth Factor Binding Proteins/pharmacology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/genetics , Oligonucleotides/pharmacology , Presynaptic Terminals/physiology , Reverse Transcriptase Polymerase Chain Reaction , Somatomedins/genetics , Synapses/physiology
9.
J Neurosci ; 18(17): 6882-91, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9712658

ABSTRACT

Increasing evidence indicates several roles for thrombin-like serine proteases and their cognate inhibitors (serpins) in normal development and/or pathology of the nervous system. In addition to its prominent role in thrombosis and/or hemostasis, thrombin inhibits neurite outgrowth in neuroblastoma and primary neuronal cells in vitro, prevents stellation of glial cells, and induces cell death in glial and neuronal cell cultures. Thrombin is known to act via a cell surface protease-activated receptor (PAR-1), and recent evidence suggests that rodent neurons express PAR-1. Previously, we have shown that the thrombin inhibitor, protease nexin-1, significantly prevents neuronal cell death both in vitro and in vivo. Here we have examined the effects of human alpha-thrombin and the presence and/or activation of PAR-1 on the survival and differentiation of highly enriched cultures of embryonic chick spinal motoneurons. We show that thrombin significantly decreased the mean neurite length, prevented neurite branching, and induced motoneuron death by an apoptosis-like mechanism in a dose-dependent manner. These effects were prevented by cotreatment with hirudin, a specific thrombin inhibitor. Treatment of the cultures with a synthetic thrombin receptor-activating peptide (SFLLRNP) mimicked the deleterious effects of thrombin on motoneurons. Furthermore, cotreatment of the cultures with inhibitors of caspase activities completely prevented the death of motoneurons induced by either thrombin or SFLLRNP. These findings indicate that (1) embryonic avian spinal motoneurons express functional PAR-1 and (2) activation of this receptor induces neuronal cell degeneration and death via stimulation of caspases. Together with previous reports, our results suggest that thrombin, its receptor(s), and endogenous thrombin inhibitors may be important regulators of neuronal cell fate during development, after injury, and in pathology of the nervous system.


Subject(s)
Apoptosis/drug effects , Cysteine Endopeptidases/drug effects , Motor Neurons/drug effects , Neurites/drug effects , Spinal Cord/drug effects , Thrombin/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Enzyme Activation , Enzyme Inhibitors/pharmacology , Hirudins/pharmacology , Motor Neurons/cytology , Spinal Cord/cytology
10.
J Comp Neurol ; 396(2): 158-68, 1998 Jun 29.
Article in English | MEDLINE | ID: mdl-9634139

ABSTRACT

Experimental lesions have been used widely to induce motoneuron (MN) degeneration as a model to test the ability of different trophic molecules to prevent lesion-induced alterations. However, the morphological mechanisms of spinal MN death following different types of lesions is not clear at the present time. In this study, we have characterized the morphological characteristics of MN cell death by examining DNA fragmentation and the ultrastructural and light microscopic morphological features of MNs following different types of spinal nerve injury (i.e., axotomy and avulsion) in the developing and adult mouse. In neonatal mice, axotomy induced cell death as well as the atrophy of MNs that survived the injury. DNA fragmentation could be detected by using the terminal deoxynucleotidyl transferase (TUNEL) method during the cell death process following neonatal axotomy, whereas TUNEL labeling was not observed following either neonatal or adult avulsion. However, with the exception of TUNEL labeling, the morphological characteristics of MN death following neonatal axotomy and avulsion were similar, and both resembled most closely the form of programmed cell death termed cytoplasmic or type 3B, which exhibits similarities as well as differences with currently accepted definitions of apoptosis. By contrast, adult avulsion resulted in a type of degeneration that resembled necrosis more closely. However, even there, the morphology was mixed, showing characteristics of both apoptosis and necrosis. These results indicate that the mode of MN degeneration is complex and is related to developmental age and type of lesion.


Subject(s)
Motor Neurons/pathology , Nerve Degeneration/pathology , Peripheral Nerve Injuries , Spinal Nerves/pathology , Animals , Animals, Newborn , Apoptosis/genetics , Axotomy , DNA Fragmentation , DNA Nucleotidylexotransferase , Genetic Techniques , Mice , Mice, Inbred BALB C , Microscopy, Electron , Necrosis , Spinal Nerves/growth & development
11.
Brain Res Brain Res Rev ; 25(1): 85-95, 1997 Sep 30.
Article in English | MEDLINE | ID: mdl-9370052

ABSTRACT

There is increasing evidence suggesting that members of the serine protease family, including thrombin, chymotrypsin, urokinase plasminogen activator, and kallikrein, may play a role in normal development and/or pathology of the nervous system. Serine proteases and their cognate inhibitors have been shown to be increased in the neural parenchyma and cerebrospinal fluid following injury to the blood brain barrier. Zymogen precursors of thrombin and thrombin-like proteases as well as their receptors have also been localized in several distinct regions of the developing or adult brain. Thrombin-like proteases have been shown to exert deleterious effects, including neurite retraction and death, on different neuronal and non-neuronal cell populations in vitro. These effects appear to be mediated through cell surface receptors and can be prevented or reversed with specific serine protease inhibitors (serpins). Furthermore, we have recently shown that treatment with protease nexin-1 (a serpin that inhibits thrombin-like proteases) promotes the survival and growth of spinal motoneurons during the period of programmed cell death and following injury. Taken together, these observations suggest that thrombin-like proteases play a deleterious role, whereas serpins promote the development and maintenance of neuronal cells. Thus, changes in the balance between serine proteases and their cognate inhibitors may lead to pathological states similar to those associated with some neurodegenerative diseases such as Alzheimer's disease. The present review summarizes the current state of research involving such serine proteases and speculates on the possible role of these thrombin-like proteases in the development, plasticity and pathology of the nervous system.


Subject(s)
Nervous System Diseases/physiopathology , Nervous System/physiopathology , Neuronal Plasticity , Receptors, Thrombin/metabolism , Serine Endopeptidases/metabolism , Thrombin/metabolism , Animals , Brain/pathology , Brain/physiology , Brain/physiopathology , Brain Diseases/pathology , Brain Diseases/physiopathology , Humans , Nervous System/pathology , Nervous System Diseases/pathology , Nervous System Physiological Phenomena , Neurons/pathology , Neurons/physiology , Spinal Cord/pathology , Spinal Cord/physiology , Spinal Cord/physiopathology
12.
J Comp Neurol ; 381(3): 353-72, 1997 May 12.
Article in English | MEDLINE | ID: mdl-9133573

ABSTRACT

Neuromuscular transmission and muscle activity during early stages of embryonic development are known to influence the differentiation and survival of motoneurons and to affect interactions with their muscle targets. We have examined neuromuscular development in an avian genetic mutant, crooked neck dwarf (cn/cn), in which a major phenotype is the chronic absence of the spontaneous, neurally mediated movements (motility) that are characteristic of avian and other vertebrate embryos and fetuses. The primary genetic defect in cn/cn embryos responsible for the absence of motility appears to be the lack of excitation-contraction coupling. Although motility in mutant embryos is absent from the onset of activity on embryonic days (E) 3-4, muscle differentiation appears histologically normal up to about E8. After E8, however, previously separate muscles fuse or coalesce secondarily, and myotubes exhibit a progressive series of histological and ultrastructural degenerative changes, including disarrayed myofibrils, dilated sarcoplasmic vesicles, nuclear membrane blebbing, mitochondrial swelling, nuclear inclusions, and absence of junctional end feet. Mutant muscle cells do not develop beyond the myotube stage, and by E18-E20 most muscles have almost completely degenerated. Prior to their breakdown and degeneration, mutant muscles are innervated and synaptic contacts are established. In fact, quantitative analysis indicates that, prior to the onset of muscle degeneration, mutant muscles are hyperinnervated. There is increased branching of motoneuron axons and an increased number of synaptic contacts in the mutant muscle on E8. Naturally occurring cell death of limb-innervating motoneurons is also significantly reduced in cn/cn embryos. Mutant embryos have 30-40% more motoneurons in the brachial and lumbar spinal cord by the end of the normal period of cell death. Electrophysiological recordings (electromyographic and direct records form muscle nerves) failed to detect any differences in the activity of control vs. mutant embryos despite the absence of muscular contractile activity in the mutant embryos. The alpha-ryanodine receptor that is genetically abnormal in homozygote cn/cn embryos is not normally expressed in the spinal cord. Taken together, these data argue against the possibility that the mutant phenotype described here is caused by the perturbation of a central nervous system (CNS)-expressed alpha-ryanodine receptor. The hyperinnervation of skeletal muscle and the reduction of motoneuron death that are observed in cn/cn embryos also occur in genetically paralyzed mouse embryos and in pharmacologically paralyzed avian and rat embryos. Because a primary common feature in all three of these models is the absence of muscle activity, it seems likely that the peripheral excitation of muscle by motoneurons during normal development is a major factor in regulating retrograde muscle-derived (or muscle-associated) signals that control motoneuron differentiation and survival.


Subject(s)
Chick Embryo/physiology , Motor Neurons/cytology , Mutation , Neuromuscular Junction/physiology , Animals , Antibody Specificity , Calcium Channels/analysis , Calcium Channels/immunology , Calmodulin-Binding Proteins/analysis , Cell Count , Cell Death/physiology , Cell Survival/physiology , Electrophysiology , Heterozygote , Microscopy, Electron , Motor Neurons/chemistry , Motor Neurons/physiology , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/analysis , Muscle Proteins/immunology , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Nervous System Diseases/physiopathology , Neuromuscular Junction/ultrastructure , Paralysis/genetics , Ryanodine Receptor Calcium Release Channel , Spinal Cord/embryology , Spinal Cord/physiopathology
13.
J Neurol Sci ; 153(1): 20-4, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9455973

ABSTRACT

Several new neurotrophic factors have been recently identified and shown to prevent motoneuron death in vitro and in vivo. One such agent is brain-derived neurotrophic factor (BDNF). In this study, we tested BDNF on an animal model of early-onset motoneuron disease: the paralysé mouse mutant, characterized by a progressive skeletal muscle atrophy and the loss of 30-35% of spinal lumbar motoneurons between the first and second week post-natal. The results show that subcutaneous injections of 1 or 10 mg/kg BDNF did not have any significant effect in increasing the mean survival time of mutant mice or in preventing the loss of motor function and total body weight in paralysé mice. The weight and choline acetyltransferase activity of specific muscles and the number of motoneurons in the spinal cords were identical in BDNF-treated and placebo-injected paralysé mice. These results suggest that BDNF does not act on the disease process in paralysé mice in the conditions we used. By contrast, BDNF has previously been shown to partially prevent the loss of motor function in the wobbler mouse, a suggested model of later-onset motoneuron disease. Taken together these findings suggest that BDNF acts differently on early and late-onset motoneuron diseases. It is however possible that treatment of paralysé mice with BDNF or combinations of different neurotrophic factors prior to the phenotypical expression of the paralysé mutation may prevent the loss of motor function and motoneurons in mutant mice.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Motor Neuron Disease/genetics , Motor Neuron Disease/prevention & control , Neuromuscular Diseases/genetics , Neuromuscular Diseases/prevention & control , Paralysis/genetics , Animals , Body Weight/drug effects , Choline O-Acetyltransferase/metabolism , Mice , Mice, Neurologic Mutants , Motor Neurons/drug effects , Postural Balance/drug effects , Survival Analysis
14.
Cell Tissue Res ; 286(2): 219-23, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8854890

ABSTRACT

During normal development of many vertebrate species, substantial numbers of neurons in the central and peripheral nervous system undergo naturally occurring (or programmed) cell death. For example, approximately 50% of spinal motoneurons degenerate and die at a time when these cells are establishing synaptic connections with their target muscles in the chick, mouse, rat, and human. It is generally thought that the survival of developing motoneurons depends on access to trophic molecules. Motoneurons that survive the period of programmed cell death may also die following injury in the developing or adult animal. Increasing evidence suggests that glial-cell-line-derived neurotrophic factor (GDNF) plays a physiological and/or pharmacological role in the survival of various neuronal cell types, including motoneurons. In this paper, we review the survival and growth-promoting effects of GDNF on spinal motoneurons during the period of programmed cell death and following injury.


Subject(s)
Motor Neurons/drug effects , Nerve Growth Factors , Nerve Tissue Proteins/pharmacology , Neuroprotective Agents/pharmacology , Spinal Cord/cytology , Animals , Axons/physiology , Cell Death/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Chick Embryo , Glial Cell Line-Derived Neurotrophic Factor , Mice , Mice, Inbred BALB C , Motor Neurons/cytology , Motor Neurons/ultrastructure , Nerve Degeneration/drug effects , Rhizotomy , Spinal Cord/embryology , Spinal Cord/surgery
15.
Brain Res Dev Brain Res ; 96(1-2): 204-9, 1996 Oct 23.
Article in English | MEDLINE | ID: mdl-8922682

ABSTRACT

The platelet-derived growth factor receptor alpha subunit (PDGFR alpha) is expressed by glial precursors, glial cells, and some peripheral neurons during normal rodent development. Its ligands are expressed ubiquitously in neurons, including sensory and motor neurons. Thus, neuronally secreted PDGF-A may play a paracrine role in the development of both glial cells and peripheral neurons. The Patch (Ph) mutation, which is a deletion of the PDGFR alpha, is a homozygous embryonic lethal mutation in the mouse. Previously, several developmental abnormalities, including deficiencies in connective tissues in many organs, aberrant neural crest cell migration, and defects in non-neuronal derivatives of crest cells, have been shown to be associated with the Patch mutation. However, whether and the extent to which motor and sensory neurons are affected by the mutation are not known. Here, we have examined the survival and/or morphological differentiation of spinal motor and sensory (dorsal root ganglion) neurons during the period of naturally occurring cell death, i.e., between E14 and E18, in control and Ph/Ph mice. The results show a 65-70% decrease in motor and sensory neuron numbers in Ph/Ph mice, compared to controls, at all stages examined. Furthermore, motoneurons in Ph/Ph mice were significantly smaller than those in controls. Because of the bidirectional nature of neuron-glial cell interactions, these results suggest that PDGFR alpha plays an important role in glial cell development and, thus, indirectly in neuronal cell development or, alternatively, that PDGF and the PDGFR alpha are directly involved in peripheral neuron survival and development by an autocrine/paracrine mechanism.


Subject(s)
Motor Neurons/physiology , Neurons, Afferent/physiology , Peptide Fragments/genetics , Receptors, Platelet-Derived Growth Factor/genetics , Spinal Cord/embryology , Animals , Cell Count , Cell Differentiation/physiology , Cell Size , Cell Survival/physiology , Embryonic and Fetal Development/physiology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Neurologic Mutants , Receptors, Platelet-Derived Growth Factor/chemistry , Spinal Cord/cytology
16.
Cell Stress Chaperones ; 1(3): 161-6, 1996 Sep.
Article in English | MEDLINE | ID: mdl-9222601

ABSTRACT

Elevation of intracellular heat shock protein (Hsp)70 increases resistance of cells to many physical and metabolic insults. We tested the hypothesis that treatment with Hsc70 can also produce that effect, using the model of axotomy-induced neuronal death in the neonatal mouse. The sciatic nerve was sectioned and in some animals purified bovine brain Hsc70 was applied to the proximal end of the nerve immediately thereafter and again 3 days later. Seven days postaxotomy, the surviving sensory neurons of the lumbar dorsal root ganglion (DRG) and motoneurons of the lumbar ventral spinal cord were counted to assess cell death. Axotomy induced the death of approximately 33% of DRG neurons and 50% of motoneurons, when examined 7 days postinjury. Application of exogenous Hsc70 prevented axotomy-induced death of virtually all sensory neurons, but did not significantly alter motoneuron death. Thus, Hsc70 may prove to be useful in the repair of peripheral sensory nerve damage.


Subject(s)
Axons/physiology , Cell Death/drug effects , Ganglia, Spinal/drug effects , HSP70 Heat-Shock Proteins/pharmacology , Motor Neurons/drug effects , Animals , Animals, Newborn , Ganglia, Spinal/cytology , Mice , Motor Neurons/cytology
17.
J Neuropathol Exp Neurol ; 55(6): 698-703, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8642395

ABSTRACT

The survival and morphometric characteristics of lumbar spinal motoneurons were examined in the paralysé mouse mutant. Affected (par/par) mice can be first recognized at approximately postnatal day (PN) 7 to 8 and are characterized by their smaller-than-normal body size, a progressive generalized muscle weakness, and lack of coordination. Mutant mice die by PN16-18, when they have become almost completely paralyzed. Previously, we have shown that this mutation involves alteration of several developmental aspects of the neuromuscular system. However, whether ventral (or anterior) horn motoneurons degenerate and die during the course of the disease was unknown. We report here that at the time the mutant phenotype can be first identified (i.e. approximately PN8), lumbar motoneuron numbers in the lateral motor column of the spinal cord of paralysé mice were not significantly different from those of control littermates. In contrast, by PN14, there was a significant (30 to 35%) decrease in motoneuron numbers in mutant compared to control mice. Furthermore, motoneuron (nuclear and soma) sizes were significantly decreased in the mutants at both stages examined, i.e. PN8 and PN14. These results show that the paralysé mutation involves atrophy and subsequent death of anterior horn motoneurons. Together with the rapid progression and the severity of the disease, these results suggest that the paralysé mouse may represent a good animal model for studying early-onset human motor neuron diseases such as spinal muscular atrophy.


Subject(s)
Mice, Mutant Strains/physiology , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Nerve Degeneration/genetics , Animals , Body Weight/physiology , Cell Death/genetics , Disease Models, Animal , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Motor Neurons/physiology , Muscle Weakness/pathology , Muscular Atrophy, Spinal/pathology , Mutation/physiology , Phenotype
18.
Proc Natl Acad Sci U S A ; 92(21): 9771-5, 1995 Oct 10.
Article in English | MEDLINE | ID: mdl-7568215

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has been shown to rescue developing motoneurons in vivo and in vitro from both naturally occurring and axotomy-induced cell death. To test whether GDNF has trophic effects on adult motoneurons, we used a mouse model of injury-induced adult motoneuron degeneration. Injuring adult motoneuron axons at the exit point of the nerve from the spinal cord (avulsion) resulted in a 70% loss of motoneurons by 3 weeks following surgery and a complete loss by 6 weeks. Half of the loss was prevented by GDNF treatment. GDNF also induced an increase (hypertrophy) in the size of surviving motoneurons. These data provide strong evidence that the survival of injured adult mammalian motoneurons can be promoted by a known neurotrophic factor, suggesting the potential use of GDNF in therapeutic approaches to adult-onset motoneuron diseases such as amyotrophic lateral sclerosis.


Subject(s)
Motor Neurons/physiology , Nerve Degeneration/drug effects , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/pharmacology , Spinal Nerve Roots/physiology , Animals , Brain-Derived Neurotrophic Factor , Cell Count , Cell Line , Glial Cell Line-Derived Neurotrophic Factor , Histocytochemistry , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred BALB C , Motor Neurons/cytology , Motor Neurons/drug effects , NADPH Dehydrogenase/isolation & purification , Nitric Oxide Synthase/isolation & purification , Rats , Rats, Sprague-Dawley , Spinal Nerve Roots/cytology , Spinal Nerve Roots/drug effects , Spinal Nerve Roots/surgery
19.
Exp Neurol ; 134(1): 49-55, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7672037

ABSTRACT

We have examined the in vivo survival effect of ciliary neurotrophic factor (CNTF) on sensory, i.e., dorsal root ganglion (DRG) neurons during the period of naturally occurring (programmed) cell death and following axotomy in the developing chick and mouse. Administration of CNTF during the period of naturally occurring cell death, from Embryonic Day (E) 6 to E10 in the chick and E14 to E18 in the mouse, had no significant effect in preventing the death of DRG neurons in either species. Axotomy on E12 in the chick or on Postnatal Day (PN) 5 in the mouse resulted in a 60% and a 33% decrease, respectively, in ipsilateral DRG neuron numbers by E16 (chick) or by PN12 (mouse), when compared to contralateral controls. CNTF treatment prevented axotomy-induced cell death of DRG neurons in both the chick and mouse. Daily administration of CNTF following axotomy in E12 chicks significantly increased (72%) DRG neurons by E16. Similarly, CNTF completely rescued mouse DRG neurons from axotomy-induced death. These results show that although CNTF has no effect on naturally occurring death of chick or mouse sensory neurons, this agent has significant ability to rescue sensory neurons following axotomy. These findings suggest that CNTF may be an effective therapeutic agent for the prevention of injury-induced death of vertebrate sensory neurons.


Subject(s)
Apoptosis , Denervation , Ganglia, Spinal/embryology , Nerve Tissue Proteins/pharmacology , Neurons, Afferent/physiology , Animals , Animals, Newborn/physiology , Cell Survival/drug effects , Chick Embryo , Ciliary Neurotrophic Factor , Embryonic and Fetal Development , Lumbosacral Region , Mice/embryology , Mice, Inbred BALB C , Nerve Growth Factors/pharmacology , Neurons, Afferent/drug effects , Sciatic Nerve/embryology
20.
Arch Histol Cytol ; 58(2): 139-49, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7576866

ABSTRACT

For nearly 70 years apoptosis has been known to be a form of cell death distinct from necrosis as well as an important regressive event during the normal development of the nervous system. For example, in the chick, mouse, rat and human approximately 50% of postmitotic neurons die naturally during embryonic or fetal development. It is generally accepted that neurons die during this period by apoptosis. After the period of naturally occurring cell death, the surviving neurons may undergo degeneration and death due to injury or disease later either during development or in adulthood. Recently, apoptosis has been suggested to be involved in the abnormal neuronal death that occurs following axonal injury or in neurodegenerative diseases such as amyotrophic lateral sclerosis and Alzheimer's. Although little is known about the etiology of these diseases, progress is steadily being made toward understanding their underlying mechanisms. For diseases of spinal motoneurons, during the past two years gene mutations have been identified in patients with familial amyotrophic lateral sclerosis or spinal muscular atrophy. Furthermore, a number of in vitro, in vivo, and mutant animal models have been developed in order to study the factors which control motoneuron survival and/or death. Here, we review the morphological differences between necrotic and apoptotic cell death and some of the methods used to differentiate the two pathways. We also discuss motoneuron cell death during development, following injury and in disease, and its prevention by different agents, including neurotrophic factors.


Subject(s)
Apoptosis/physiology , Nervous System Physiological Phenomena , Animals , Humans , Models, Biological , Necrosis
SELECTION OF CITATIONS
SEARCH DETAIL
...