Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Neuroanat ; 16: 937596, 2022.
Article in English | MEDLINE | ID: mdl-35958734

ABSTRACT

Sexual phenotypic differences in the nervous system are one of the most prevalent features across the animal kingdom. The molecular mechanisms responsible for sexual dimorphism throughout metazoan nervous systems are extremely diverse, ranging from intrinsic cell autonomous mechanisms to gonad-dependent endocrine control of sexual traits, or even extrinsic environmental cues. In recent years, the DMRT ancient family of transcription factors has emerged as being central in the development of sex-specific differentiation in all animals in which they have been studied. In this review, we provide an overview of the function of Dmrt genes in nervous system sexual regulation from an evolutionary perspective.

2.
Elife ; 112022 03 01.
Article in English | MEDLINE | ID: mdl-35230240

ABSTRACT

Axons of the corpus callosum (CC) mediate the interhemispheric communication required for complex perception in mammals. In the somatosensory (SS) cortex, the CC exchanges inputs processed by the primary (S1) and secondary (S2) areas, which receive tactile and pain stimuli. During early postnatal life, a multistep process involving axonal navigation, growth, and refinement, leads to precise CC connectivity. This process is often affected in neurodevelopmental disorders such as autism and epilepsy. We herein show that in mice, expression of the axonal signaling receptor Neuropilin 1 (Nrp1) in SS layer (L) 2/3 is temporary and follows patterns that determine CC connectivity. At postnatal day 4, Nrp1 expression is absent in the SS cortex while abundant in the motor area, creating a sharp border. During the following 3 weeks, Nrp1 is transiently upregulated in subpopulations of SS L2/3 neurons, earlier and more abundantly in S2 than in S1. In vivo knock-down and overexpression experiments demonstrate that transient expression of Nrp1 does not affect the initial development of callosal projections in S1 but is required for subsequent S2 innervation. Moreover, knocking-down Nrp1 reduces the number of S2L2/3 callosal neurons due to excessive postnatal refinement. Thus, an exquisite temporal and spatial regulation of Nrp1 expression determines SS interhemispheric maps.


Subject(s)
Axons/physiology , Corpus Callosum/cytology , Neurons/physiology , Neuropilin-1/metabolism , Somatosensory Cortex/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neuropilin-1/genetics , Somatosensory Cortex/cytology , Somatosensory Cortex/embryology
3.
Curr Opin Neurobiol ; 68: 159-166, 2021 06.
Article in English | MEDLINE | ID: mdl-33915498

ABSTRACT

While single-cell transcriptomics in the brain has uncovered a vast diversity of neural cell types in unprecedented detail, it is becoming increasingly urgent to address what exactly their functional roles are in the context of circuits and behavior. In this review, we discuss the molecular profiling of cell types in circuits underlying social behaviors in mice as a prominent case study. We first highlight key roles of molecularly identified sensory and downstream neurons involved in sexually dimorphic behaviors. We then propose future opportunities to define cell types using multimodal criteria, especially gene expression, physiology, as well as the developmental origin, to advance our understanding of these circuits.


Subject(s)
Sex Characteristics , Sexual Behavior, Animal , Animals , Brain , Mice , Neurons , Social Behavior
4.
Genetics ; 215(3): 665-681, 2020 07.
Article in English | MEDLINE | ID: mdl-32444379

ABSTRACT

We explore here the cis-regulatory logic that dictates gene expression in specific cell types in the nervous system. We focus on a set of eight genes involved in the synthesis, transport, and breakdown of three neurotransmitter systems: acetylcholine (unc-17/VAChT, cha-1/ChAT, cho-1/ChT, and ace-2/AChE), glutamate (eat-4/VGluT), and γ-aminobutyric acid (unc-25/GAD, unc-46/LAMP, and unc-47/VGAT). These genes are specifically expressed in defined subsets of cells in the nervous system. Through transgenic reporter gene assays, we find that the cellular specificity of expression of all of these genes is controlled in a modular manner through distinct cis-regulatory elements, corroborating the previously inferred piecemeal nature of specification of neurotransmitter identity. This modularity provides the mechanistic basis for the phenomenon of "phenotypic convergence," in which distinct regulatory pathways can generate similar phenotypic outcomes (i.e., the acquisition of a specific neurotransmitter identity) in different neuron classes. We also identify cases of enhancer pleiotropy, in which the same cis-regulatory element is utilized to control gene expression in distinct neuron types. We engineered a cis-regulatory allele of the vesicular acetylcholine transporter, unc-17/VAChT, to assess the functional contribution of a "shadowed" enhancer. We observed a selective loss of unc-17/VAChT expression in one cholinergic pharyngeal pacemaker motor neuron class and a behavioral phenotype that matches microsurgical removal of this neuron. Our analysis illustrates the value of understanding cis-regulatory information to manipulate gene expression and control animal behavior.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Neurons/metabolism , Neurotransmitter Agents/metabolism , Regulatory Sequences, Nucleic Acid , Vesicular Acetylcholine Transport Proteins/genetics , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Genetic Pleiotropy , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Neurons/classification , Neurotransmitter Agents/genetics , Vesicular Acetylcholine Transport Proteins/metabolism , Vesicular Glutamate Transport Proteins/genetics , Vesicular Glutamate Transport Proteins/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
5.
Wiley Interdiscip Rev Dev Biol ; 9(4): e374, 2020 07.
Article in English | MEDLINE | ID: mdl-32012462

ABSTRACT

One approach to understand the construction of complex systems is to investigate whether there are simple design principles that are commonly used in building such a system. In the context of nervous system development, one may ask whether the generation of its highly diverse sets of constituents, that is, distinct neuronal cell types, relies on genetic mechanisms that share specific common features. Specifically, are there common patterns in the function of regulatory genes across different neuron types and are those regulatory mechanisms not only used in different parts of one nervous system, but are they conserved across animal phylogeny? We address these questions here by focusing on one specific, highly conserved and well-studied regulatory factor, the POU homeodomain transcription factor UNC-86. Work over the last 30 years has revealed a common and paradigmatic theme of unc-86 function throughout most of the neuron types in which Caenorhabditis elegans unc-86 is expressed. Apart from its role in preventing lineage reiterations during development, UNC-86 operates in combination with distinct partner proteins to initiate and maintain terminal differentiation programs, by coregulating a vast array of functionally distinct identity determinants of specific neuron types. Mouse orthologs of unc-86, the Brn3 genes, have been shown to fulfill a similar function in initiating and maintaining neuronal identity in specific parts of the mouse brain and similar functions appear to be carried out by the sole Drosophila ortholog, Acj6. The terminal selector function of UNC-86 in many different neuron types provides a paradigm for neuronal identity regulation across phylogeny. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Invertebrate Organogenesis > Worms Nervous System Development > Vertebrates: Regional Development.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , POU Domain Factors/genetics , Transcription Factor Brn-3C/genetics , Animals , Caenorhabditis elegans/classification , Caenorhabditis elegans/cytology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation , Cell Lineage/genetics , Conserved Sequence , Drosophila Proteins/metabolism , Drosophila melanogaster/classification , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nervous System/cytology , Nervous System/growth & development , Nervous System/metabolism , Neurons/cytology , Neurons/metabolism , POU Domain Factors/metabolism , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Transcription Factor Brn-3C/metabolism
6.
Genetics ; 214(1): 163-178, 2020 01.
Article in English | MEDLINE | ID: mdl-31776169

ABSTRACT

Members of the superfamily of solute carrier (SLC) transmembrane proteins transport diverse substrates across distinct cellular membranes. Three SLC protein families transport distinct neurotransmitters into synaptic vesicles to enable synaptic transmission in the nervous system. Among them is the SLC17A6/7/8 family of vesicular glutamate transporters, which endows specific neuronal cell types with the ability to use glutamate as a neurotransmitter. The genome of the nematode Caenorhabditis elegans encodes three SLC17A6/7/8 family members, one of which, eat-4/VGLUT, has been shown to be involved in glutamatergic neurotransmission. Here, we describe our analysis of the two remaining, previously uncharacterized SLC17A6/7/8 family members, vglu-2 and vglu-3 These two genes directly neighbor one another and are the result of a recent gene duplication event in C. elegans, but not in other Caenorhabditis species. Compared to EAT-4, the VGLU-2 and VGLU-3 protein sequences display a more distant similarity to canonical, vertebrate VGLUT proteins. We tagged both genomic loci with gfp and detected no expression of vglu-3 at any stage of development in any cell type of both C. elegans sexes. In contrast, vglu-2::gfp is dynamically expressed in a restricted set of distinct cell types. Within the nervous system, vglu-2::gfp is exclusively expressed in a single interneuron class, AIA, where it localizes to vesicular structures in the soma, but not along the axon, suggesting that VGLU-2 may not be involved in synaptic transport of glutamate. Nevertheless, vglu-2 mutants are partly defective in the function of the AIA neuron in olfactory behavior. Outside the nervous system, VGLU-2 is expressed in collagen secreting skin cells where VGLU-2 most prominently localizes to early endosomes, and to a lesser degree to apical clathrin-coated pits, the trans-Golgi network, and late endosomes. On early endosomes, VGLU-2 colocalizes most strongly with the recycling promoting factor SNX-1, a retromer component. Loss of vglu-2 affects the permeability of the collagen-containing cuticle of the worm, and based on the function of a vertebrate VGLUT1 protein in osteoclasts, we speculate that vglu-2 may have a role in collagen trafficking in the skin. We conclude that C. elegans SLC17A6/7/8 family members have diverse functions within and outside the nervous system.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Biological Transport , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Genome , Humans , Phylogeny , Sequence Homology , Synaptic Transmission , Vesicular Glutamate Transport Proteins/genetics
7.
Elife ; 82019 01 01.
Article in English | MEDLINE | ID: mdl-30599092

ABSTRACT

The molecular mechanisms that control the timing of sexual differentiation in the brain are poorly understood. We found that the timing of sexually dimorphic differentiation of postmitotic, sex-shared neurons in the nervous system of the Caenorhabditis elegans male is controlled by the temporally regulated miRNA let-7 and its target lin-41, a translational regulator. lin-41 acts through lin-29a, an isoform of a conserved Zn finger transcription factor, expressed in a subset of sex-shared neurons only in the male. Ectopic lin-29a is sufficient to impose male-specific features at earlier stages of development and in the opposite sex. The temporal, sexual and spatial specificity of lin-29a expression is controlled intersectionally through the lin-28/let-7/lin-41 heterochronic pathway, sex chromosome configuration and neuron-type-specific terminal selector transcription factors. Two Doublesex-like transcription factors represent additional sex- and neuron-type specific targets of LIN-41 and are regulated in a similar intersectional manner.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Cell Differentiation/genetics , MicroRNAs/genetics , Nervous System/metabolism , Neurons/metabolism , Transcription Factors/genetics , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Larva/metabolism , Male , Nervous System/cytology , Sequence Homology, Amino Acid , Sex Differentiation/genetics , Sex Factors , Time Factors , Transcription Factors/metabolism
8.
Curr Biol ; 28(17): 2813-2823.e2, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30146154

ABSTRACT

Many distinct regulatory factors have been shown to be required for the proper initiation of neuron-type-specific differentiation programs, but much less is known about the regulatory programs that maintain the differentiated state in the adult [1-3]. One possibility is that regulatory factors that initiate a terminal differentiation program during development are continuously required to maintain the differentiated state. Here, we test this hypothesis by investigating the function of two orthologous POU homeobox genes in nematodes and mice. The C. elegans POU homeobox gene unc-86 is a terminal selector that is required during development to initiate the terminal differentiation program of several distinct neuron classes [4-13]. Through post-developmental removal of unc-86 activity, we show here that unc-86 is also continuously required throughout the life of many neuron classes to maintain neuron-class-specific identity features. Similarly, the mouse unc-86 ortholog Brn3a/POU4F1 has been shown to control the initiation of the terminal differentiation program of distinct neuron types across the mouse brain, such as the medial habenular neurons [14-20]. By conditionally removing Brn3a in the adult mouse central nervous system, we show that, like its invertebrate ortholog unc-86, Brn3a is also required for the maintenance of terminal identity features of medial habenular neurons. In addition, Brn3a is required for the survival of these neurons, indicating that identity maintenance and survival are genetically linked. We conclude that the continuous expression of transcription factors is essential for the active maintenance of the differentiated state of a neuron across phylogeny.


Subject(s)
Caenorhabditis elegans/genetics , Cell Differentiation/physiology , Neurons/physiology , POU Domain Factors/metabolism , Animals , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genes, Homeobox , Mice , Mitosis , Neurogenesis , POU Domain Factors/genetics , Tamoxifen/pharmacology , Transcription Factors/metabolism
9.
Genetics ; 206(3): 1251-1269, 2017 07.
Article in English | MEDLINE | ID: mdl-28684604

ABSTRACT

The nervous system of most animals is sexually dimorphic but such dimorphisms are generally poorly mapped on an anatomical, cellular, and molecular level. The adult nervous system of the nematode Caenorhabditis elegans displays a number of clearly defined anatomical sexual dimorphisms, but molecular features of sexually dimorphic neurons remain sparse. In this resource paper, we provide a comprehensive atlas of neurotransmitters used in the nervous system of the male and compare it to that of the hermaphrodite. Among the three major neurotransmitter systems, acetylcholine (ACh) is the most frequently used, followed by glutamate (Glu), and lastly γ-aminobutyric acid (GABA). Many male-specific neurons utilize multiple neurotransmitter systems. Interestingly, we find that neurons that are present in both sexes alter their neurotransmitter usage depending on the sex of the animal. One neuron scales up its usage of ACh, another becomes serotonergic in males, and another one adds a new neurotransmitter (glutamate) to its nonsex-specific transmitter (ACh). In all these cases, neurotransmitter changes are correlated with substantial changes in synaptic connectivity. We assembled the neurotransmitter maps of the male-specific nervous system into a comprehensive atlas that describes the anatomical position of all the neurons of the male-specific nervous system relative to the sex-shared nervous system. We exemplify the usefulness of the neurotransmitter atlas by using it as a tool to define the expression pattern of a synaptic organizer molecule in the male tail. Taken together, the male neurotransmitter atlas provides an entry point for future functional and developmental analysis of the male nervous system.


Subject(s)
Acetylcholine/metabolism , Caenorhabditis elegans/metabolism , Glutamic Acid/metabolism , Serotonin/metabolism , Sex Characteristics , Synapses/metabolism , Animals , Caenorhabditis elegans/physiology , Female , Male , Nervous System/cytology , Nervous System/metabolism , Neurons/classification , Neurons/metabolism , Synapses/classification , Synaptic Transmission
10.
Curr Biol ; 27(2): 199-209, 2017 Jan 23.
Article in English | MEDLINE | ID: mdl-28065609

ABSTRACT

Functional and anatomical sexual dimorphisms in the brain are either the result of cells that are generated only in one sex or a manifestation of sex-specific differentiation of neurons present in both sexes. The PHC neuron pair of the nematode C. elegans differentiates in a strikingly sex-specific manner. In hermaphrodites the PHC neurons display a canonical pattern of synaptic connectivity similar to that of other sensory neurons, but in males PHC differentiates into a densely connected hub sensory neuron/interneuron, integrating a large number of male-specific synaptic inputs and conveying them to both male-specific and sex-shared circuitry. We show that the differentiation into such a hub neuron involves the sex-specific scaling of several components of the synaptic vesicle machinery, including the vesicular glutamate transporter eat-4/VGLUT, induction of neuropeptide expression, changes in axonal projection morphology, and a switch in neuronal function. We demonstrate that these molecular and anatomical remodeling events are controlled cell autonomously by the phylogenetically conserved Doublesex homolog dmd-3, which is both required and sufficient for sex-specific PHC differentiation. Cellular specificity of dmd-3 action is ensured by its collaboration with non-sex-specific terminal selector-type transcription factors, whereas the sex specificity of dmd-3 action is ensured by the hermaphrodite-specific transcriptional master regulator of hermaphroditic cell identity tra-1, which represses the transcription of dmd-3 in hermaphrodite PHC. Taken together, our studies provide mechanistic insights into how neurons are specified in a sexually dimorphic manner.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Cell Differentiation , Neurons/cytology , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Female , Gene Expression Regulation, Developmental , Male , Neurons/physiology , Sex Characteristics , Sex Differentiation , Synapses/physiology , Transcription Factors/genetics
11.
Elife ; 42015 Dec 25.
Article in English | MEDLINE | ID: mdl-26705699

ABSTRACT

Nervous system maps are of critical importance for understanding how nervous systems develop and function. We systematically map here all cholinergic neuron types in the male and hermaphrodite C. elegans nervous system. We find that acetylcholine (ACh) is the most broadly used neurotransmitter and we analyze its usage relative to other neurotransmitters within the context of the entire connectome and within specific network motifs embedded in the connectome. We reveal several dynamic aspects of cholinergic neurotransmitter identity, including a sexually dimorphic glutamatergic to cholinergic neurotransmitter switch in a sex-shared interneuron. An expression pattern analysis of ACh-gated anion channels furthermore suggests that ACh may also operate very broadly as an inhibitory neurotransmitter. As a first application of this comprehensive neurotransmitter map, we identify transcriptional regulatory mechanisms that control cholinergic neurotransmitter identity and cholinergic circuit assembly.


Subject(s)
Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/physiology , Cholinergic Fibers , Connectome , Nervous System/anatomy & histology , Acetylcholine/metabolism , Animals , Cholinergic Agents/metabolism , Female , Interneurons , Male , Neurotransmitter Agents/metabolism
12.
Development ; 142(14): 2464-77, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26153233

ABSTRACT

Neurogenesis involves deeply conserved patterning molecules, such as the proneural basic helix-loop-helix transcription factors. Sox proteins and specifically members of the SoxB and SoxC groups are another class of conserved transcription factors with an important role in neuronal fate commitment and differentiation in various species. In this study, we examine the expression of all five Sox genes of the nematode C. elegans and analyze the effect of null mutant alleles of all members of the SoxB and SoxC groups on nervous system development. Surprisingly, we find that, unlike in other systems, neither of the two C. elegans SoxB genes sox-2 (SoxB1) and sox-3 (SoxB2), nor the sole C. elegans SoxC gene sem-2, is broadly expressed throughout the embryonic or adult nervous system and that all three genes are mostly dispensable for embryonic neurogenesis. Instead, sox-2 is required to maintain the developmental potential of blast cells that are generated in the embryo but divide only postembryonically to give rise to differentiated neuronal cell types. Moreover, sox-2 and sox-3 have selective roles in the terminal differentiation of specific neuronal cell types. Our findings suggest that the common themes of SoxB gene function across phylogeny lie in specifying developmental potential and, later on, in selectively controlling terminal differentiation programs of specific neuron types, but not in broadly controlling neurogenesis.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/embryology , Gene Expression Regulation, Developmental , Nervous System/embryology , Neurogenesis/physiology , Neurons/cytology , SOXB1 Transcription Factors/physiology , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation , Cell Lineage , Male , Motor Neurons/metabolism , Mutation , SOXC Transcription Factors/physiology , Signal Transduction , Transgenes
13.
Cell ; 155(3): 659-73, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243022

ABSTRACT

The choice of using one of many possible neurotransmitter systems is a critical step in defining the identity of an individual neuron type. We show here that the key defining feature of glutamatergic neurons, the vesicular glutamate transporter EAT-4/VGLUT, is expressed in 38 of the 118 anatomically defined neuron classes of the C. elegans nervous system. We show that distinct cis-regulatory modules drive expression of eat-4/VGLUT in distinct glutamatergic neuron classes. We identify 13 different transcription factors, 11 of them homeodomain proteins, that act in distinct combinations in 25 different glutamatergic neuron classes to initiate and maintain eat-4/VGLUT expression. We show that the adoption of a glutamatergic phenotype is linked to the adoption of other terminal identity features of a neuron, including cotransmitter phenotypes. Examination of mouse orthologs of these homeodomain proteins resulted in the identification of mouse LHX1 as a regulator of glutamatergic neurons in the brainstem.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Homeodomain Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Receptors, Glutamate/metabolism , Animals , Caenorhabditis elegans/metabolism , Mice , Neurons/classification , Sensory Receptor Cells/metabolism , Transcription Factors/metabolism , Vesicular Glutamate Transport Proteins
14.
J Neurosci ; 31(24): 8894-904, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21677173

ABSTRACT

Despite extensive research to develop an effective neuroprotective strategy for the treatment of ischemic stroke, therapeutic options remain limited. Although caspase-dependent death is thought to play a prominent role in neuronal injury, direct evidence of active initiator caspases in stroke and the functional relevance of this activity have not previously been shown. Using an unbiased caspase-trapping technique in vivo, we isolated active caspase-9 from ischemic rat brain within 1 h of reperfusion. Pathogenic relevance of active caspase-9 was shown by intranasal delivery of a novel cell membrane-penetrating highly specific inhibitor for active caspase-9 at 4 h postreperfusion (hpr). Caspase-9 inhibition provided neurofunctional protection and established caspase-6 as its downstream target. The temporal and spatial pattern of expression demonstrates that neuronal caspase-9 activity induces caspase-6 activation, mediating axonal loss by 12 hpr followed by neuronal death within 24 hpr. Collectively, these results support selective inhibition of these specific caspases as an effective therapeutic strategy for stroke.


Subject(s)
Caspase 6/physiology , Enzyme Inhibitors/therapeutic use , Infarction, Middle Cerebral Artery , Inhibitor of Apoptosis Proteins/therapeutic use , Nervous System Diseases , Neurons/pathology , Administration, Intranasal , Aldehydes/pharmacology , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Caspase 6/deficiency , Caspase 9/metabolism , Caspase Inhibitors , Cysteine Proteinase Inhibitors/therapeutic use , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , In Vitro Techniques , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Nervous System Diseases/pathology , PTEN Phosphohydrolase/chemistry , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/therapeutic use , Rats , Rats, Wistar , Time Factors
15.
Biochem J ; 415(2): 165-82, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18800967

ABSTRACT

Dysregulation of life and death at the cellular level leads to a variety of diseases. In the nervous system, aberrant neuronal death is an outstanding feature of neurodegenerative diseases. Since the discovery of the caspase family of proteases, much effort has been made to determine how caspases function in disease, including neurodegenerative diseases. Although many papers have been published examining caspases in neuronal death and disease, the pathways have not been fully clarified. In the present review, we examine the potential players in the death pathways, the current tools for examining these players and the models for studying neurological disease. Alzheimer's disease, the most common neurodegenerative disorder, and cerebral ischaemia, the most common cause of neurological death, are used to illustrate our current understanding of death signalling in neurodegenerative diseases. A better understanding of the neuronal death pathways would provide targets for the development of therapeutic interventions for these diseases.


Subject(s)
Apoptosis/physiology , Nervous System Diseases/physiopathology , Neurons/cytology , Signal Transduction , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Caspases/metabolism , Humans , Models, Biological , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neurons/metabolism
16.
J Neurovirol ; 12(6): 420-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17162658

ABSTRACT

Herpes simplex virus type 1 (HSV-1) expresses an immediate-early protein, ICP47, that blocks the major histocompatibility complex class I antigen presentation pathway by binding to the transporter associated with antigen presentation (TAP). The result is the virus' evasion of the immune system. Although the interaction between ICP47 and TAP has been examined in vitro, this paper is the first to report their interaction in vivo. In C57BL/6 adult female mice, ICP47-defective virus (Delta ICP47, F strain) was less able to invade the organs studied than was wild-type HSV-1 F strain, showing that ICP47 influences general invasiveness. However, the neuroinvasiveness of the Delta ICP47 virus was recovered in TAP-deficient mice, indicating that the TAP-ICP47 interaction is specific to neural tissues. HSV-1 F strain showed no significant differences in their invasiveness in TAP-deficient and wild-type mice. Therefore, although ICP47 appears to be essential for invasion, the presence of TAP appears not to be crucial. Western blotting showed TAP1 expression to increase by at least fourfold in the brains and adrenal glands of infected mice. This suggests that TAP plays an important role in the host defense system. This increased expression may be particularly important in the encephalon since the baseline protein levels of this organ are low (ratio adrenal protein level/encephalon protein level > 100). However, Delta ICP47 virus provoked no significant increase in the brain TAP1 levels of wild-type mice because it could not invade this organ. These results suggest that ICP47 plays a role in infection, and that TAP1 production is regulated during viral challenge.


Subject(s)
Antigens, Ly/metabolism , Brain/virology , Herpesvirus 1, Human/pathogenicity , Immediate-Early Proteins/metabolism , Membrane Proteins/metabolism , Viral Proteins/metabolism , Animals , Antigens, Ly/immunology , Blotting, Western , Female , Herpesvirus 1, Human/immunology , Immediate-Early Proteins/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...