Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
J Frailty Aging ; 6(1): 37-45, 2017.
Article in English | MEDLINE | ID: mdl-28244557

ABSTRACT

This systematic review summarizes the psychometric properties of goal-setting instruments that are applied within geriatric rehabilitation. PubMed Medline and Embase were systematically searched for eligible articles. Studies were included if they were conducted in a somatic or neurological rehabilitation setting, included patients aged ≥55 years and provided data on instruments' psychometric properties (validity, reliability, responsiveness), utility and/or feasibility. Eleven studies were included. Seven studies, all conducted by one research group, evaluated Goal-Attainment Scaling (GAS), two studies assessed the Canadian Occupational Performance Measure (COPM) and one study the Self-Identified Goals Assessment (SIGA), which is based on the COPM. One study assessed a core set of the International Classification of Functioning, Disability and Health (ICF) framework. High concurrent, content and predictive validity and inter-rater reliability were found for GAS. Responsiveness appears to be excellent. Concurrent validity and inter-rater reliability of the COPM and content validity of both the COPM and SIGA appear to be good. Responsiveness of both instruments seems to be poor. Content validity of the ICF core set was found to be fair; responsiveness appears to be very poor. There is little published data on goal-setting instruments in geriatric rehabilitation. Evidence for its psychometric properties may support GAS as goal-setting instrument and additional outcome measure. However, more research is required in order to evaluate GAS, as research conducted in other health care settings may provide important additional findings. Before the COPM (or SIGA) can be recommended as goal-setting instrument, its psychometric properties require further research.


Subject(s)
Health Services for the Aged , Patient Care Planning , Psychometrics , Rehabilitation/organization & administration , Aged , Health Services for the Aged/organization & administration , Health Services for the Aged/standards , Humans , Middle Aged , Outcome Assessment, Health Care , Psychometrics/methods , Psychometrics/standards , Treatment Outcome
2.
Oncogene ; 25(7): 1081-9, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16247468

ABSTRACT

Focal adhesion kinase (FAK) is a critical component in transducing signals downstream of both integrins and growth factor receptors. To determine how the loss of FAK affects the epidermis in vivo, we have generated a mouse model with a keratinocyte-restricted deletion of fak (FAKK5 KO mice). FAK(K5 KO) mice displayed three major phenotypes--irregularities of hair cycle, sebaceous glands hypoplasia, and a thinner epidermis--pointing to defects in the proliferative capacity of multipotent stem cells found in the bulge. FAK-null keratinocytes in conventional primary culture undergo massive apoptosis hindering further analyses, whereas the defects observed in vivo do not shorten the mouse lifespan. These results suggest that the structure and the signaling environment of the native tissue may overcome the lack of signaling through FAK. Our findings point to the importance of in vivo and three-dimensional in vitro models in analyses of cell migration, proliferation, and survival. Surprisingly, the difference between FAKloxP/+ and FAKK5 KO mice in wound closure was not statistically significant, suggesting that in vivo loss of FAK does not affect migration/proliferation of basal keratinocytes in the same way as it affects multipotent stem cells of the skin.


Subject(s)
Focal Adhesion Kinase 1/genetics , Hair/abnormalities , Keratinocytes/enzymology , Wound Healing , Animals , Cell Movement , Cell Proliferation , Epidermal Cells , Epidermis/abnormalities , Epidermis/growth & development , Female , Focal Adhesion Kinase 1/deficiency , Gene Deletion , Hair/cytology , Hair/growth & development , Keratin-15 , Keratin-5 , Keratinocytes/cytology , Keratins/metabolism , Male , Mice , Mice, Knockout , Sebaceous Glands/abnormalities , Sebaceous Glands/cytology , Wound Healing/genetics
3.
Exp Eye Res ; 76(1): 125-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12589782

ABSTRACT

Rod photoreceptors are susceptible to light-induced cell death. Previous results have suggested that the neurotrophin receptor p75 in Müller cells controls photoreceptor cell death during light-exposure by suppressing trophic factor release; and consequently, if p75 is blocked or eliminated during light-exposure, apoptosis is delayed. We explored this question by examining photoreceptor cell survival in albino p75(-/-) mice as well as their heterozygous and homozygous littermates. Photoreceptor cell death was examined in semi-thin sections by counting the remaining rows of photoreceptors. No difference in the amount of cell death was found between p75(+/+) and p75(-/-) animals, whereas the single copy of p75 in the heterozygous p75(+/-) mice provided significant neuroprotection. Cell death in the wild-type animals may indeed be mediated by p75, whereas other known apoptosis pathways may be activated in the p75(-/-) mice. The pro-apoptotic activity of the p75 receptor may have been partially suppressed in the heterozygous p75(+/-) mice by the silencing effect of the Trk receptor. Thus, our results suggest that p75 signaling does not mediate the main apoptosis pathway activated during light-damage.


Subject(s)
Apoptosis/radiation effects , Light/adverse effects , Radiation Injuries/pathology , Receptors, Nerve Growth Factor/physiology , Retinal Rod Photoreceptor Cells/radiation effects , Animals , Mice , Mice, Mutant Strains , Receptor, Nerve Growth Factor
4.
J Neurobiol ; 53(2): 143-56, 2002 Nov 05.
Article in English | MEDLINE | ID: mdl-12382272

ABSTRACT

The development and evolution of the inner ear sensory patches and their innervation is reviewed. Recent molecular developmental data suggest that development of these sensory patches is a developmental recapitulation of the evolutionary history. These data suggest that the ear generates multiple, functionally diverse sensory epithelia by dividing a single sensory primordium. Those epithelia will establish distinct identities through the overlapping expression of genes of which only a few are currently known. One of these distinctions is the unique pattern of hair cell polarity. A hypothesis is presented on how the hair cell polarity may relate to the progressive segregation of the six sensory epithelia. Besides being markers for sensory epithelia development, neurotrophins are also expressed in delaminating cells that migrate toward the developing vestibular and cochlear ganglia. These delaminating cells originate from multiple sites at or near the developing sensory epithelia and some also express neuronal markers such as NeuroD. The differential origin of precursors raises the possibility that some sensory neurons acquire positional information before they delaminate the ear. Such an identity of these delaminating sensory neurons may be used both to navigate their dendrites to the area they delaminated from, as well as to help them navigate to their central target. The navigational properties of sensory neurons as well as the acquisition of discrete sensory patch phenotypes implies a much more sophisticated subdivision of the developing otocyst than the few available gene expression studies suggest.


Subject(s)
Cochlea/embryology , Cochlea/innervation , Animals , Body Patterning , Cell Differentiation , Cell Lineage , Cochlea/metabolism , Embryonic Induction/genetics , Epithelium/embryology , Epithelium/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Neurons, Afferent/cytology , Polysaccharides/biosynthesis
5.
Exp Neurol ; 172(1): 81-91, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11681842

ABSTRACT

During early postnatal development, apoptosis of retinal ganglion cells (RGCs) is regulated by target contact with the optic tectum. The neurotrophins BDNF and NT-4, but not NGF, prevent the apoptosis of retinal ganglion cells that is otherwise observed after target ablation or axotomy. Thus receptors activated by BDNF and NT-4 are candidates to mediate the early postnatal survival of RGCs. BDNF and NT-4, but not NGF, bind to all isoforms of the receptor TrkB, whether or not they contain a tyrosine kinase domain. To examine the roles of TrkB receptor isoforms in early postnatal survival, we compared RGC numbers in wild-type mice to those in a mutant lacking all isoforms of TrkB. Surprisingly, no reduction in RGCs was observed in the mutant at postnatal day 16, the latest age at which these animals are consistently viable, so TrkB signaling is not essential for target-dependent survival of these cells. In wild-type mice, RGCs also are lost gradually during adulthood, possibly due to oxidative stress. To determine whether TrkB signaling regulates this phase of RGC degeneration, RGC numbers were examined in a viable mutant of TrkB that expresses only about 25% the normal level of TrkB receptor kinase. Compared to controls, approximately 20% of the RGC were lost in mutant 3-month-old-animals. Thus, TrkB signaling is not required for survival of RGCs during the period of target-dependent survival, but does appear to reduce degeneration of RGCs in adult animals.


Subject(s)
Receptor, trkB/metabolism , Retinal Ganglion Cells/metabolism , Aging/metabolism , Animals , Axons/ultrastructure , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Count , Cell Survival/physiology , Heterozygote , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, trkB/deficiency , Receptor, trkB/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/cytology , Retina/growth & development , Retinal Ganglion Cells/cytology , Signal Transduction/physiology , Superior Colliculi/cytology , beta-Galactosidase/genetics
6.
Development ; 128(13): 2421-32, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11493560

ABSTRACT

The POU domain transcription factors Brn3a, Brn3b and Brn3c are required for the proper development of sensory ganglia, retinal ganglion cells, and inner ear hair cells, respectively. We have investigated the roles of Brn3a in neuronal differentiation and target innervation in the facial-stato-acoustic ganglion. We show that absence of Brn3a results in a substantial reduction in neuronal size, abnormal neuronal migration and downregulation of gene expression, including that of the neurotrophin receptor TrkC, parvalbumin and Brn3b. Selective loss of TrkC neurons in the spiral ganglion of Brn3a(-/-) cochlea leads to an innervation defect similar to that of TrkC(-/-) mice. Most remarkably, our results uncover a novel role for Brn3a in regulating axon pathfinding and target field innervation by spiral and vestibular ganglion neurons. Loss of Brn3a results in severe retardation in development of the axon projections to the cochlea and the posterior vertical canal as early as E13.5. In addition, efferent axons that use the afferent fibers as a scaffold during pathfinding also show severe misrouting. Interestingly, despite the well-established roles of ephrins and EphB receptors in axon pathfinding, expression of these molecules does not appear to be affected in Brn3a(-/-) mice. Thus, Brn3a must control additional downstream genes that are required for axon pathfinding.


Subject(s)
Axons/physiology , DNA-Binding Proteins/metabolism , Geniculate Ganglion/cytology , Spiral Ganglion/cytology , Transcription Factors/metabolism , Vestibular Nerve/cytology , Animals , Cell Differentiation , Cell Size , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Ear, Inner/cytology , Gene Expression Regulation , Mice , Mice, Mutant Strains , Neurons, Afferent/cytology , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphB4 , Receptors, Eph Family , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transcription Factor Brn-3B , Transcription Factor Brn-3C , Transcription Factors/genetics , Transcription Factors/physiology
7.
J Neurosci ; 21(16): 6170-80, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11487640

ABSTRACT

Previous work suggested qualitatively different effects of neurotrophin 3 (NT-3) in cochlear innervation patterning in different null mutants. We now show that all NT-3 null mutants have a similar phenotype and lose all neurons in the basal turn of the cochlea. To understand these longitudinal deficits in neurotrophin mutants, we have compared the development of the deficit in the NT-3 mutant to the spatial-temporal expression patterns of brain-derived neurotrophic factor (BDNF) and NT-3, using lacZ reporters in each gene and with expression of the specific neurotrophin receptors, trkB and trkC. In the NT-3 mutant, almost normal numbers of spiral ganglion neurons form, but fiber outgrowth to the basal turn is eliminated by embryonic day (E) 13.5. Most neurons are lost between E13.5 and E15.5. During the period preceding apoptosis, NT-3 is expressed in supporting cells, whereas BDNF is expressed mainly in hair cells, which become postmitotic in an apical to basal temporal gradient. During the period of neuronal loss, BDNF is absent from the basal cochlea, accounting for the complete loss of basal turn neurons in the NT-3 mutant. The spatial gradients of neuronal loss in these two mutants appear attributable to spatial-temporal gradients of neurotrophin expression. Our immunocytochemical data show equal expression of their receptors, TrkB and TrkC, in spiral sensory neurons and thus do not relate to the basal turn loss. Mice in which NT-3 was replaced by BDNF show a qualitative normal pattern of innervation at E13.5. This suggests that the pattern of expression of neurotrophins rather than their receptors is essential for the spatial loss of spiral sensory neurons in NT-3 null mutants.


Subject(s)
Cochlea/innervation , Cochlea/metabolism , Gene Expression Regulation, Developmental , Neurotrophin 3/biosynthesis , Neurotrophin 3/genetics , Afferent Pathways/cytology , Afferent Pathways/embryology , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Cell Count , Cell Survival/genetics , Cochlea/embryology , Genes, Reporter , Heterozygote , Homozygote , Immunohistochemistry , Lac Operon , Mice , Mice, Mutant Strains , Mutation , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Phenotype , Receptor, trkB/biosynthesis , Receptor, trkC/biosynthesis , Spiral Ganglion/cytology , Spiral Ganglion/embryology
8.
Annu Rev Neurosci ; 24: 677-736, 2001.
Article in English | MEDLINE | ID: mdl-11520916

ABSTRACT

Neurotrophins regulate development, maintenance, and function of vertebrate nervous systems. Neurotrophins activate two different classes of receptors, the Trk family of receptor tyrosine kinases and p75NTR, a member of the TNF receptor superfamily. Through these, neurotrophins activate many signaling pathways, including those mediated by ras and members of the cdc-42/ras/rho G protein families, and the MAP kinase, PI-3 kinase, and Jun kinase cascades. During development, limiting amounts of neurotrophins function as survival factors to ensure a match between the number of surviving neurons and the requirement for appropriate target innervation. They also regulate cell fate decisions, axon growth, dendrite pruning, the patterning of innervation and the expression of proteins crucial for normal neuronal function, such as neurotransmitters and ion channels. These proteins also regulate many aspects of neural function. In the mature nervous system, they control synaptic function and synaptic plasticity, while continuing to modulate neuronal survival.


Subject(s)
Nerve Growth Factors/physiology , Neurons/physiology , Animals , Humans , Ion Channels/physiology , MAP Kinase Signaling System/physiology , Models, Neurological , Receptors, Nerve Growth Factor/physiology , Signal Transduction/physiology , Vertebrates
9.
J Cell Biol ; 154(2): 447-58, 2001 Jul 23.
Article in English | MEDLINE | ID: mdl-11470831

ABSTRACT

The epithelial-mesenchymal interactions required for kidney organogenesis are disrupted in mice lacking the integrin alpha8beta1. None of this integrin's known ligands, however, appears to account for this phenotype. To identify a more relevant ligand, a soluble integrin alpha8beta1 heterodimer fused to alkaline phosphatase (AP) has been used to probe blots and cDNA libraries. In newborn mouse kidney extracts, alpha8beta1-AP detects a novel ligand of 70-90 kD. This protein, named nephronectin, is an extracellular matrix protein with five EGF-like repeats, a mucin region containing a RGD sequence, and a COOH-terminal MAM domain. Integrin alpha8beta1 and several additional RGD-binding integrins bind nephronectin. Nephronectin mRNA is expressed in the ureteric bud epithelium, whereas alpha8beta1 is expressed in the metanephric mesenchyme. Nephronectin is localized in the extracellular matrix in the same distribution as the ligand detected by alpha8beta1-AP and forms a complex with alpha8beta1 in vivo. Thus, these results strongly suggest that nephronectin is a relevant ligand mediating alpha8beta1 function in the kidney. Nephronectin is expressed at numerous sites outside the kidney, so it may also have wider roles in development. The approaches used here should be generally useful for characterizing the interactions of novel extracellular matrix proteins identified through genomic sequencing projects.


Subject(s)
Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Kidney/metabolism , Receptors, Antigen/metabolism , Alkaline Phosphatase/genetics , Animals , Animals, Newborn , Base Sequence , Cloning, Molecular , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/chemistry , Humans , K562 Cells , Kidney/embryology , Ligands , Macromolecular Substances , Mesoderm/metabolism , Mice , Molecular Sequence Data , Oligopeptides/metabolism , Organ Specificity , Protein Binding/physiology , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Ureter/embryology , Ureter/metabolism
10.
Curr Opin Neurobiol ; 11(3): 272-80, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11399424

ABSTRACT

The four mammalian neurotrophins - NGF, BDNF, NT-3 and NT-4 - each bind and activate one or more of the Trk family of receptor tyrosine kinases. Through these receptors, neurotrophins activate many intracellular signaling pathways, including those controlled by Ras, the Cdc42/Rac/RhoG protein family, MAPK, PI3K and PLC-gamma, thereby affecting both development and function of the nervous system. During the past two years, several novel signaling pathways controlled by Trk receptors have been characterized, and it has become clear that membrane transport and sorting controls Trk-receptor-mediated signaling because key intermediates are localized to different membrane compartments. Three-dimensional structures of the Trk receptors, in one instance in association with a neurotrophin, have revealed the structural bases underlying specificity in neurotrophin signaling.


Subject(s)
Nerve Growth Factors/physiology , Receptors, Nerve Growth Factor/physiology , Signal Transduction/physiology , Animals , Antigens, CD/physiology , Apoptosis , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Endocytosis , Humans , Isoenzymes/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mammals/physiology , Mice , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/physiology , Neurons/physiology , Organ Specificity , PC12 Cells , Phosphatidylinositol 3-Kinases/physiology , Phospholipase C gamma , Rats , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type II , Signal Transduction/drug effects , Structure-Activity Relationship , Type C Phospholipases/physiology , ras Proteins/physiology
11.
J Neurosci ; 21(10): 3492-502, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11331378

ABSTRACT

Neuronal growth factors regulate the survival of neurons by their survival and death-promoting activity on distinct populations of neurons. The neurotrophins nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) promote neuronal survival via tyrosine kinase (Trk) receptors, whereas NGF and BDNF can also induce apoptosis in developing neurons through p75(NTR) receptors in the absence of their respective Trk receptors. Using mutant mice and inactivation of neurotrophins and their receptors with antibodies in rats, we show that endogenous NT-3 induces death of adult BDNF-dependent, axotomized corticospinal neurons (CSNs). When NT-3 is neutralized, the neurons survive even without BDNF, suggesting complete antagonism. Whereas virtually all unlesioned and axotomized CSNs express both trkB and trkC mRNA, p75 is barely detectable in unlesioned CSNs but strongly upregulated in axotomized CSNs by day 3 after lesion, the time point when cell death occurs. Blocking either cortical TrkC or p75(NTR) receptors alone prevents death, indicating that the opposing actions of NT-3 and BDNF require their respective Trk receptors, but induction of death depends on p75(NTR) cosignaling. The results show that neuronal survival can be regulated antagonistically by neurotrophins and that neurotrophins can induce neuronal death in the adult mammalian CNS. We further present evidence that signaling of tyrosine kinase receptors of the trk family can be crucially involved in the promotion of neuronal death in vivo.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Neurons/metabolism , Neurotrophin 3/physiology , Pyramidal Tracts/metabolism , Animals , Antibodies, Blocking/administration & dosage , Axotomy , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/pharmacology , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Drug Antagonism , Female , Gene Expression/drug effects , Heterozygote , Immunohistochemistry , Infusions, Parenteral , Male , Mice , Mice, Mutant Strains , Neurons/drug effects , Neurotrophin 3/antagonists & inhibitors , Neurotrophin 3/pharmacology , Pyramidal Tracts/anatomy & histology , Pyramidal Tracts/drug effects , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/genetics , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/drug effects
12.
Development ; 128(4): 471-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11171331

ABSTRACT

Cell death in the developing retina is regulated, but so far little is known about what factors regulate the cell death. Several neurotrophic factors and receptors, including the neurotrophins and Trk receptors, are expressed during the critical time. We have studied the developing avian retina with respect to the role of nerve growth factor (NGF) in these processes. Our starting point for the work was that NGF and its receptor TrkA are expressed in a partially overlapping pattern in the inner nuclear layer of the developing retina. Our results show that TrkA and NGF-expressing cells are postmitotic. The first NGF-expressing cells were found on the vitreal side of the central region of E5.5-E6 retina. This pattern changed and NGF-expressing cells identified as horizontal cells were later confined to the external inner nuclear layer. We show that these horizontal cells co-express TrkA and NGF, unlike a subpopulation of amacrine cells that only expresses TrkA. In contrast to the horizontal cells, which survive, the majority of the TrkA-expressing amacrine cells die during a period of cell death in the inner nuclear layer. Intraocular injections of NGF protein rescued the dying amacrine cells and injection of antisense oligonucleotides for NGF that block its synthesis, caused death among the TrkA-expressing horizontal cells, which normally would survive. Our results suggest that NGF supports the survival of TrkA expressing avian horizontal cells in an autocrine mode of action in the retina of E10-E12 chicks. The cells co-express TrkA and NGF and the role for NGF is to maintain the TrkA-expressing horizontal cells. The TrkA-expressing amacrine cells are not supported by NGF and subsequently die. In addition to the effect on survival, our results suggest that NGF plays a role in horizontal cell plasticity.


Subject(s)
Autocrine Communication , Cell Survival , Gene Expression Regulation, Developmental , Nerve Growth Factor/metabolism , Retina/embryology , Retina/metabolism , Animals , Bromodeoxyuridine/metabolism , Cell Death/genetics , Cell Differentiation , Cell Division , Cell Survival/drug effects , Chick Embryo , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Microinjections , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/genetics , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Retina/cytology
13.
J Biol Chem ; 276(14): 11294-301, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11150291

ABSTRACT

The TrkA receptor is activated primarily by nerve growth factor (NGF), but it can also be activated by high concentrations of neurotrophin 3 (NT-3). The pan-neurotrophin receptor p75(NTR) strongly inhibits activation of TrkA by NT-3 but not by NGF. To examine the role of p75(NTR) in regulating the specificity of TrkA signaling, we expressed both receptors in Xenopus oocytes. Application of NGF or NT-3 to oocytes expressing TrkA alone resulted in efflux of (45)Ca(2+) by a phospholipase C-gamma-dependent pathway. Coexpression of p75(NTR) with TrkA inhibited (45)Ca(2+) efflux in response to NT-3 but not NGF. The inhibitory effect on NT-3 activation of TrkA increased with increasing expression of p75(NTR). Coexpression of a truncated p75(NTR) receptor lacking all but the first 9 amino acids of the cytoplasmic domain inhibited NT-3 stimulation of (45)Ca(2+) efflux, whereas coexpression of an epidermal growth factor receptor/p75(NTR) chimera (extracellular domain of epidermal growth factor receptor with transmembrane and cytoplasmic domains of p75(NTR)) did not inhibit NT-3 signaling through TrkA. These studies demonstrated that the extracellular domain of p75(NTR) was necessary to inhibit NT-3 signaling through TrkA. Remarkably, p75(NTR) binding to NT-3 was not required to prevent signaling through TrkA, since occupying p75(NTR) with brain-derived neurotrophic factor or anti-p75 antibody (REX) did not rescue the ability of NT-3 to activate (45)Ca(2+) efflux. These data suggested a physical association between TrkA and p75(NTR). Documenting this physical interaction, we showed that p75(NTR) and TrkA could be coimmunoprecipitated from Xenopus oocytes. Our results suggest that the interaction of these two receptors on the cell surface mediated the inhibition of NT-3-activated signaling through TrkA.


Subject(s)
Neurotrophin 3/metabolism , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Humans , Neurotrophin 3/chemistry , Receptor, Nerve Growth Factor , Receptor, trkA/chemistry , Receptors, Nerve Growth Factor/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Xenopus laevis
14.
Development ; 128(3): 417-26, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11152640

ABSTRACT

A key factor in the genetically programmed development of the nervous system is the death of massive numbers of neurons. Therefore, genetic mechanisms governing cell survival are of fundamental importance to developmental neuroscience. We report that inner ear sensory neurons are dependent on a basic helix-loop-helix transcription factor called NeuroD for survival during differentiation. Mice lacking NeuroD protein exhibit no auditory evoked potentials, reflecting a profound deafness. DiI fiber staining, immunostaining and cell death assays reveal that the deafness is due to the failure of inner ear sensory neuron survival during development. The affected inner ear sensory neurons fail to express neurotrophin receptors, TrkB and TrkC, suggesting that the ability of NeuroD to support neuronal survival may be directly mediated through regulation of responsiveness to the neurotrophins.


Subject(s)
Cochlea/growth & development , Deafness/genetics , Gene Deletion , Hair Cells, Auditory, Inner/pathology , Nerve Tissue Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Death , Cell Movement , Cell Survival , Cochlea/innervation , Cochlea/pathology , Cochlea/ultrastructure , Deafness/physiopathology , Evoked Potentials, Auditory/genetics , Evoked Potentials, Auditory/physiology , Gene Expression Regulation, Developmental , Genes, Reporter , Hair Cells, Auditory, Inner/growth & development , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/ultrastructure , Helix-Loop-Helix Motifs , Histocytochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Mice, Knockout , Microscopy, Electron , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Neural Pathways/growth & development , Neural Pathways/pathology , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptor, trkC/genetics , Receptor, trkC/metabolism
15.
J Neurosci ; 20(18): 6888-97, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10995833

ABSTRACT

The neurotrophin BDNF has been shown to modulate long-term potentiation (LTP) at Schaffer collateral-CA1 hippocampal synapses. Mutants in the BDNF receptor gene trkB and antibodies to its second receptor p75NTR have been used to determine the receptors and cells involved in this response. Inhibition of p75NTR does not detectably reduce LTP or affect presynaptic function, but analyses of newly generated trkB mutants implicate TrkB. One mutant has reduced expression in a normal pattern of TrkB throughout the brain. The second mutant was created by cre-loxP-mediated removal of TrkB in CA1 pyramidal neurons of this mouse. Neither mutant detectably impacts survival or morphology of hippocampal neurons. TrkB reduction, however, affects presynaptic function and reduces the ability of tetanic stimulation to induce LTP. Postsynaptic glutamate receptors are not affected by TrkB reduction, indicating that BDNF does not modulate plasticity through postsynaptic TrkB. Consistent with this, elimination of TrkB in postsynaptic neurons does not affect LTP. Moreover, normal LTP is generated in the mutant with reduced TrkB by a depolarization-low-frequency stimulation pairing protocol that puts minimal demands on presynaptic terminal function. Thus, BDNF appears to act through TrkB presynaptically, but not postsynaptically, to modulate LTP.


Subject(s)
Hippocampus/metabolism , Long-Term Potentiation/genetics , Presynaptic Terminals/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkB/metabolism , Animals , Antigens, Differentiation/metabolism , Axons/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Hippocampus/cytology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics , Patch-Clamp Techniques , Pyramidal Cells/metabolism , RNA, Messenger/biosynthesis , Receptor, Nerve Growth Factor/antagonists & inhibitors , Receptor, trkB/deficiency , Receptor, trkB/genetics , Receptors, Glutamate/metabolism , Signal Transduction/genetics , Stem Cells
16.
J Neurophysiol ; 84(1): 472-83, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10899220

ABSTRACT

Brain-derived neurotrophic factor (BDNF) can potentiate synaptic release at newly developed frog neuromuscular junctions. Although this potentiation depends on extracellular Ca(2+) and reflects changes in acetylcholine release, little is known about the intracellular transduction or calcium signaling pathways. We have developed a video assay for neurotrophin-induced potentiation of myocyte twitching as a measure of potentiation of synaptic activity. We use this assay to show that BDNF-induced synaptic potentiation is not blocked by cadmium, indicating that Ca(2+) influx through voltage-gated Ca(2+) channels is not required. TrkB autophosphorylation is not blocked in Ca(2+)-free conditions, indicating that TrkB activity is not Ca(2+) dependent. Additionally, an inhibitor of phospholipase C interferes with BDNF-induced potentiation. These results suggest that activation of the TrkB receptor activates phospholipase C to initiate intracellular Ca(2+) release from stores which subsequently potentiates transmitter release.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Calcium/metabolism , Motor Neurons/physiology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Action Potentials/physiology , Animals , Cadmium/pharmacology , Calcium/pharmacokinetics , Calcium Channels/physiology , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Indoles/pharmacology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Oocytes/physiology , PC12 Cells , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , Phosphotyrosine/analysis , Pyrrolidinones/pharmacology , Rats , Receptor, trkB/physiology , Synapses/enzymology , Thapsigargin/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Tyrosine/metabolism , Xenopus
17.
Curr Opin Neurobiol ; 10(3): 392-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10851180

ABSTRACT

Many constituents of Wnt signaling pathways are expressed in the developing and mature nervous systems. Recent work has shown that Wnt signaling controls initial formation of the neural plate and many subsequent patterning decisions in the embryonic nervous system, including formation of the neural crest. Wnt signaling continues to be important at later stages of development. Wnts have been shown to regulate the anatomy of the neuronal cytoskeleton and the differentiation of synapses in the cerebellum. Wnt signaling has been demonstrated to regulate apoptosis and may participate in degenerative processes leading to cell death in the aging brain.


Subject(s)
Brain/cytology , Brain/growth & development , Neurons/physiology , Proto-Oncogene Proteins/physiology , Zebrafish Proteins , Aging/physiology , Animals , Brain Chemistry/physiology , Signal Transduction/physiology , Wnt Proteins
18.
Neuron ; 26(1): 233-45, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10798407

ABSTRACT

To examine functions of TrkB in the adult CNS, TrkB has been removed from neurons expressing CaMKII, primarily pyramidal neurons, using Cre-mediated recombination. A floxed trkB allele was designed so that neurons lacking TrkB express tau-beta-galactosidase. Following trkB deletion in pyramidal cells, their dendritic arbors are altered, and cortical layers II/III and V are compressed, after which there is an apparent loss of mutant neurons expressing the transcription factor SCIP but not of those expressing Otx-1. Loss of neurons expressing SCIP requires deletion of trkB within affected neurons; reduction of neuronal ER81 expression does not, suggesting both direct and indirect effects of TrkB loss. Thus, TrkB is required for the maintenance of specific populations of cells in the adult neocortex.


Subject(s)
Neocortex/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Receptor, trkB/metabolism , beta-Galactosidase/metabolism , Animals , Cell Count , DNA-Binding Proteins/metabolism , Dendrites/metabolism , Dendrites/pathology , Mice , Mice, Transgenic , Mutation/genetics , Neocortex/pathology , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/pathology , Receptor, trkB/genetics , Transcription Factors/metabolism
19.
J Neurosci ; 19(20): 8919-30, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516311

ABSTRACT

Brain-derived neurotrophic factor (BDNF) acts through TrkB, a receptor with kinase activity, and mitigates light-induced apoptosis in adult mouse rod photoreceptors. To determine whether TrkB signaling is necessary for rod development and function, we examined the retinas of mice lacking all isoforms of the TrkB receptor. Rod migration and differentiation occur in the mutant retina, but proceed at slower rates than in wild-type mice. In postnatal day 16 (P16) mutants, rod outer segment dimensions and rhodopsin content are comparable with those of photoreceptors in P12 wild type (WT). Quantitative analyses of the photoreceptor component in the electroretinogram (ERG) indicate that the gain and kinetics of the rod phototransduction signal in dark-adapted P16 mutant and P12 WT retinas are similar. In contrast to P12 WT, however, the ERG in mutant mice entirely lacks a b-wave, indicating a failure of signal transmission in the retinal rod pathway. In the inner retina of mutant mice, although cells appear anatomically and immunohistochemically normal, they fail to respond to prolonged stroboscopic illumination with the normal expression of c-fos. Absence of the b-wave and failure of c-fos expression, in view of anatomically normal inner retinal cells, suggest that lack of TrkB signaling causes a defect in synaptic signaling between rods and inner retinal cells. Retinal pigment epithelial cells and cells in the inner retina, including Müller, amacrine, and retinal ganglion cells, express the TrkB receptor, but rod photoreceptors do not. Moreover, inner retinal cells respond to exogenous BDNF with c-fos expression and extracellular signal-regulated kinase phosphorylation. Thus, interactions of rods with TrkB-expressing cells must be required for normal rod development.


Subject(s)
Aging/physiology , Receptor, trkB/physiology , Retina/physiology , Retinal Rod Photoreceptor Cells/growth & development , Synaptic Transmission/physiology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Electroretinography , Immunohistochemistry , Light , Mice , Mice, Inbred ICR , Mice, Knockout/genetics , Mutation/physiology , Proto-Oncogene Proteins c-fos/metabolism , Receptor, trkB/genetics , Reference Values , Retina/cytology , Retina/drug effects , Retina/radiation effects , Rhodopsin/metabolism
20.
Dev Biol ; 210(2): 411-27, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10357900

ABSTRACT

Superior cervical ganglia of postnatal mice with a targeted disruption of the gene for neurotrophin-3 have 50% fewer neurons than those of wild-type mice. In culture, neurotrophin-3 increases the survival of proliferating sympathetic precursors. Both precursor death (W. ElShamy et al., 1996, Development 122, 491-500) and, more recently, neuronal death (S. Wyatt et al., 1997, EMBO J. 16, 3115-3123) have been described in mice lacking NT-3. Consistent with the second report, we found that, in vivo, neurogenesis and precursor survival were unaffected by the absence of neurotrophin-3 but neuronal survival was compromised so that only 50% of the normal number of neurons survived to birth. At the time of neuron loss, neurotrophin-3 expression, assayed with a lacZ reporter, was detected in sympathetic target tissues and blood vessels, including those along which sympathetic axons grow, suggesting it may act as a retrograde neurotrophic factor, similar to nerve growth factor. To explore this possibility, we compared neuron loss in neurotrophin-3-deficient mice with that in nerve growth factor-deficient mice and found that neuronal losses occurred at approximately the same time in both mutants, but were less severe in mice lacking neurotrophin-3. Eliminating one or both neurotrophin-3 alleles in mice that lack nerve growth factor does not further reduce sympathetic neuron number in the superior cervical ganglion at E17.5 but does alter axon outgrowth and decrease salivary gland innervation. Taken together these results suggest that neurotrophin-3 is required for survival of some sympathetic neurons that also require nerve growth factor.


Subject(s)
Nerve Growth Factors/physiology , Neurons/cytology , Stellate Ganglion/embryology , Stem Cells/cytology , Superior Cervical Ganglion/embryology , Animals , Animals, Newborn , Cell Division , Cell Survival , Embryonic and Fetal Development , Genes, Reporter , Gestational Age , Mice , Mice, Knockout , Mice, Transgenic , Mitotic Index , Morphogenesis , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Neurons/physiology , Neurotrophin 3 , Stellate Ganglion/cytology , Stellate Ganglion/physiology , Stem Cells/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...