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










Publication year range
1.
Nat Commun ; 15(1): 467, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38212606

ABSTRACT

Dietary restriction (DR) delays aging, but the mechanism remains unclear. We identified polymorphisms in mtd, the fly homolog of OXR1, which influenced lifespan and mtd expression in response to DR. Knockdown in adulthood inhibited DR-mediated lifespan extension in female flies. We found that mtd/OXR1 expression declines with age and it interacts with the retromer, which regulates trafficking of proteins and lipids. Loss of mtd/OXR1 destabilized the retromer, causing improper protein trafficking and endolysosomal defects. Overexpression of retromer genes or pharmacological restabilization with R55 rescued lifespan and neurodegeneration in mtd-deficient flies and endolysosomal defects in fibroblasts from patients with lethal loss-of-function of OXR1 variants. Multi-omic analyses in flies and humans showed that decreased Mtd/OXR1 is associated with aging and neurological diseases. mtd/OXR1 overexpression rescued age-related visual decline and tauopathy in a fly model. Hence, OXR1 plays a conserved role in preserving retromer function and is critical for neuronal health and longevity.


Subject(s)
Aging , Nervous System Diseases , Humans , Female , Aging/genetics , Longevity/genetics , Neurons/metabolism , Nervous System Diseases/metabolism , Brain/metabolism , Caloric Restriction , Mitochondrial Proteins/metabolism
3.
Geroscience ; 44(4): 2195-2211, 2022 08.
Article in English | MEDLINE | ID: mdl-35381951

ABSTRACT

Elevated serum urate (hyperuricemia) promotes crystalline monosodium urate tissue deposits and gout, with associated inflammation and increased mortality. To identify modifiers of uric acid pathologies, we performed a fly Genome-Wide Association Study (GWAS) on purine metabolites using the Drosophila Genetic Reference Panel strains. We tested the candidate genes using the Drosophila melanogaster model of hyperuricemia and uric acid crystallization ("concretion formation") in the kidney-like Malpighian tubule. Medusa (mda) activity increased urate levels and inflammatory response programming. Conversely, whole-body mda knockdown decreased purine synthesis precursor phosphoribosyl pyrophosphate, uric acid, and guanosine levels; limited formation of aggregated uric acid concretions; and was sufficient to rescue lifespan reduction in the fly hyperuricemia and gout model. Levels of mda homolog FAM214A were elevated in inflammatory M1- and reduced in anti-inflammatory M2-differentiated mouse bone marrow macrophages, and influenced intracellular uric acid levels in human HepG2 transformed hepatocytes. In conclusion, mda/FAM214A acts in a conserved manner to regulate purine metabolism, promotes disease driven by hyperuricemia and associated tissue inflammation, and provides a potential novel target for uric acid-driven pathologies.


Subject(s)
Drosophila Proteins , Gout , Hyperuricemia , Animals , Humans , Mice , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Genome-Wide Association Study , Gout/genetics , Gout/complications , Gout/metabolism , Hyperuricemia/genetics , Hyperuricemia/complications , Hyperuricemia/metabolism , Inflammation/genetics , Inflammation/complications , Purines/metabolism , Uric Acid/urine , Drosophila Proteins/genetics
4.
Database (Oxford) ; 20202020 12 31.
Article in English | MEDLINE | ID: mdl-33382884

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death worldwide for all genders and across most racial and ethnic groups. However, different races and ethnicities exhibit different rates of CVD and its related cardiorenal and metabolic comorbidities, suggesting differences in genetic predisposition and risk of onset, as well as socioeconomic and lifestyle factors (diet, exercise, etc.) that act upon an individual's unique underlying genetic background. Here, we present HeartBioPortal2.0, a major update to HeartBioPortal, the world's largest CVD genetics data precision medicine platform for harmonized CVD-relevant genetic variants, which now enables search and analysis of human genetic information related to heart disease across ethnically diverse populations and cardiovascular/renal/metabolic quantitative traits pertinent to CVD pathophysiology. HeartBioPortal2.0 is structured as a cloud-based computing platform and knowledge portal that consolidates a multitude of CVD-relevant genomic data modalities into a single powerful query and browsing interface between data and user via a user-friendly web application publicly available to the scientific research community. Since its initial release, HeartBioPortal2.0 has added new cardiovascular/renal/metabolic disease-relevant gene expression data as well as genetic association data from numerous large-scale genome-wide association study consortiums such as CARDIoGRAMplusC4D, TOPMed, FinnGen, AFGen, MESA, MEGASTROKE, UK Biobank, CHARGE, Biobank Japan and MyCode, among other studies. In addition, HeartBioPortal2.0 now includes support for quantitative traits and ethnically diverse populations, allowing users to investigate the shared genetic architecture of any gene or its variants across the continuous cardiometabolic spectrum from health (e.g. blood pressure traits) to disease (e.g. hypertension), facilitating the understanding of CVD trait genetics that inform health-to-disease transitions and endophenotypes. Custom visualizations in the new and improved user interface, including performance enhancements and new security features such as user authentication, collectively re-imagine HeartBioPortal's user experience and provide a data commons that co-locates data, storage and computing infrastructure in the context of studying the genetic basis behind the leading cause of global mortality. Database URL: https://www.heartbioportal.com/.


Subject(s)
Cardiovascular Diseases , Genome-Wide Association Study , Cardiovascular Diseases/genetics , Female , Genetic Predisposition to Disease , Genomics , Humans , Male , Phenotype
5.
Blood Adv ; 4(15): 3495-3506, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32750130

ABSTRACT

Each year, blood transfusions save millions of lives. However, under current blood-matching practices, sensitization to non-self-antigens is an unavoidable adverse side effect of transfusion. We describe a universal donor typing platform that could be adopted by blood services worldwide to facilitate a universal extended blood-matching policy and reduce sensitization rates. This DNA-based test is capable of simultaneously typing most clinically relevant red blood cell (RBC), human platelet (HPA), and human leukocyte (HLA) antigens. Validation was performed, using samples from 7927 European, 27 South Asian, 21 East Asian, and 9 African blood donors enrolled in 2 national biobanks. We illustrated the usefulness of the platform by analyzing antibody data from patients sensitized with multiple RBC alloantibodies. Genotyping results demonstrated concordance of 99.91%, 99.97%, and 99.03% with RBC, HPA, and HLA clinically validated typing results in 89 371, 3016, and 9289 comparisons, respectively. Genotyping increased the total number of antigen typing results available from 110 980 to >1 200 000. Dense donor typing allowed identification of 2 to 6 times more compatible donors to serve 3146 patients with multiple RBC alloantibodies, providing at least 1 match for 176 individuals for whom previously no blood could be found among the same donors. This genotyping technology is already being used to type thousands of donors taking part in national genotyping studies. Extraction of dense antigen-typing data from these cohorts provides blood supply organizations with the opportunity to implement a policy of genomics-based precision matching of blood.


Subject(s)
Blood Donors , Blood Transfusion , Genotype , Humans , Isoantibodies , Prospective Studies
6.
PLoS Genet ; 16(7): e1008835, 2020 07.
Article in English | MEDLINE | ID: mdl-32644988

ABSTRACT

In most organisms, dietary restriction (DR) increases lifespan. However, several studies have found that genotypes within the same species vary widely in how they respond to DR. To explore the mechanisms underlying this variation, we exposed 178 inbred Drosophila melanogaster lines to a DR or ad libitum (AL) diet, and measured a panel of 105 metabolites under both diets. Twenty four out of 105 metabolites were associated with the magnitude of the lifespan response. These included proteinogenic amino acids and metabolites involved in α-ketoglutarate (α-KG)/glutamine metabolism. We confirm the role of α-KG/glutamine synthesis pathways in the DR response through genetic manipulations. We used covariance network analysis to investigate diet-dependent interactions between metabolites, identifying the essential amino acids threonine and arginine as "hub" metabolites in the DR response. Finally, we employ a novel metabolic and genetic bipartite network analysis to reveal multiple genes that influence DR lifespan response, some of which have not previously been implicated in DR regulation. One of these is CCHa2R, a gene that encodes a neuropeptide receptor that influences satiety response and insulin signaling. Across the lines, variation in an intronic single nucleotide variant of CCHa2R correlated with variation in levels of five metabolites, all of which in turn were correlated with DR lifespan response. Inhibition of adult CCHa2R expression extended DR lifespan of flies, confirming the role of CCHa2R in lifespan response. These results provide support for the power of combined genomic and metabolomic analysis to identify key pathways underlying variation in this complex quantitative trait.


Subject(s)
Aging/genetics , Drosophila Proteins/genetics , Longevity/genetics , Metabolome/genetics , Receptors, G-Protein-Coupled/genetics , Aging/metabolism , Aging/pathology , Animals , Caloric Restriction , Diet , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/genetics , Insulin/genetics , Metabolomics , Mutation/genetics , Signal Transduction/genetics
7.
Curr Biol ; 30(14): 2749-2760.e3, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32502405

ABSTRACT

Dietary restriction (DR) is the most robust means to extend lifespan and delay age-related diseases across species. An underlying assumption in the aging field is that DR enhances both lifespan and physical activity through similar mechanisms, but this has not been rigorously tested in different genetic backgrounds. Furthermore, nutrient response genes responsible for lifespan extension or age-related decline in functionality remain underexplored in natural populations. To address this, we measured nutrient-dependent changes in lifespan and age-related decline in climbing ability in the Drosophila Genetic Reference Panel fly strains. On average, DR extended lifespan and delayed decline in climbing ability, but there was a lack of correlation between these traits across individual strains, suggesting that distinct genetic factors modulate these traits independently and that genotype determines response to diet. Only 50% of strains showed positive response to DR for both lifespan and climbing ability, 14% showed a negative response for one trait but not both, and 35% showed no change in one or both traits. Through GWAS, we uncovered a number of genes previously not known to be diet responsive nor to influence lifespan or climbing ability. We validated decima as a gene that alters lifespan and daedalus as one that influences age-related decline in climbing ability. We found that decima influences insulin-like peptide transcription in the GABA receptor neurons downstream of short neuropeptide F precursor (sNPF) signaling. Modulating these genes produced independent effects on lifespan and physical activity decline, which suggests that these age-related traits can be regulated through distinct mechanisms.


Subject(s)
Animal Nutritional Physiological Phenomena/genetics , Behavior, Animal/physiology , Diet Therapy , Drosophila/genetics , Drosophila/physiology , Genome-Wide Association Study , Insulin/metabolism , Locomotion/genetics , Longevity/genetics , Peptides/metabolism , Animals , Drosophila Proteins/metabolism , GABAergic Neurons , Genotype , Neuropeptides/metabolism , Quantitative Trait, Heritable , Signal Transduction/genetics , Signal Transduction/physiology
9.
G3 (Bethesda) ; 8(11): 3421-3431, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30194090

ABSTRACT

A feature common to late onset proteinopathic disorders is an accumulation of toxic protein conformers and aggregates in affected tissues. In the search for potential drug targets, many studies used high-throughput screens to find genes that modify the cytotoxicity of misfolded proteins. A complement to this approach is to focus on strategies that use protein aggregation as a phenotypic readout to identify pathways that control aggregate formation and maintenance. Here we use natural variation between strains of budding yeast to genetically map loci that influence the aggregation of a polyglutamine-containing protein derived from a mutant form of huntingtin, the causative agent in Huntington disease. Linkage analysis of progeny derived from a cross between wild and laboratory yeast strains revealed two polymorphic loci that modify polyglutamine aggregation. One locus contains the gene RFU1 which modifies ubiquitination states of misfolded proteins targeted by the E3-ubiquitin ligase complex Rsp5 Activity of the Rsp5 complex, and the mammalian homolog NEDD4, are critical in maintaining protein homeostasis in response to proteomic stress. Our analysis also showed linkage of the aggregation phenotype to a distinct locus containing a gene encoding the Rsp5-interacting Bul2 protein. Allele-swap experiments validated the impact of both RFU1 and BUL2 on huntingtin aggregation. Furthermore, we found that the nematode Caenorhabditis elegans' ortholog of Rsp5, wwp-1, also negatively regulates polyglutamine aggregation. Knockdown of the NEDD4 in human cells likewise altered polyglutamine aggregation. Taken together, these results implicate conserved processes involving the ubiquitin regulation network that modify protein aggregation and provide novel therapeutic targets for polyglutamine and other protein folding diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Caenorhabditis elegans Proteins/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Peptides/physiology , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Caenorhabditis elegans , Genetic Variation , HEK293 Cells , Humans , Mutation , Saccharomycetales/physiology
10.
BMC Genomics ; 17(1): 867, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27809764

ABSTRACT

BACKGROUND: Obesity-related diseases are major contributors to morbidity and mortality in the developed world. Molecular diagnostics and targets of therapies to combat nutritional imbalance are urgently needed in the clinic. Invertebrate animals have been a cornerstone of basic research efforts to dissect the genetics of metabolism and nutrient response. We set out to use fruit flies reared on restricted and nutrient-rich diets to identify genes associated with starvation resistance, body mass and composition, in a survey of genetic variation across the Drosophila Genetic Reference Panel (DGRP). RESULTS: We measured starvation resistance, body weight and composition in DGRP lines on each of two diets and used several association mapping strategies to harness this panel of phenotypes for molecular insights. We tested DNA sequence variants for a relationship with single metabolic traits and with multiple traits at once, using a scheme for cross-phenotype association mapping; we focused our association tests on homologs of human disease genes and common polymorphisms; and we tested for gene-by-diet interactions. The results revealed gene and gene-by-diet associations between 17 variants and body mass, whole-body triglyceride and glucose content, or starvation resistance. Focused molecular experiments validated the role in body mass of an uncharacterized gene, CG43921 (which we rename heavyweight), and previously unknown functions for the diacylglycerol kinase rdgA, the huntingtin homolog htt, and the ceramide synthase schlank in nutrient-dependent body mass, starvation resistance, and lifespan. CONCLUSIONS: Our findings implicate a wealth of gene candidates in fly metabolism and nutrient response, and ascribe novel functions to htt, rdgA, hwt and schlank.


Subject(s)
Drosophila/physiology , Genetic Association Studies , Nutritional Physiological Phenomena/genetics , Phenotype , Animals , Drosophila Proteins , Energy Metabolism , Female , Genetic Variation , Genotype , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Longevity/genetics , Quantitative Trait, Heritable
11.
Cell Metab ; 23(1): 143-54, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26626459

ABSTRACT

Endogenous circadian clocks orchestrate several metabolic and signaling pathways that are known to modulate lifespan, suggesting clocks as potential targets for manipulation of metabolism and lifespan. We report here that the core circadian clock genes, timeless (tim) and period (per), are required for the metabolic and lifespan responses to DR in Drosophila. Consistent with the involvement of a circadian mechanism, DR enhances the amplitude of cycling of most circadian clock genes, including tim, in peripheral tissues. Mass-spectrometry-based lipidomic analysis suggests a role of tim in cycling of specific medium chain triglycerides under DR. Furthermore, overexpression of tim in peripheral tissues improves its oscillatory amplitude and extends lifespan under ad libitum conditions. Importantly, effects of tim on lifespan appear to be mediated through enhanced fat turnover. These findings identify a critical role for specific clock genes in modulating the effects of nutrient manipulation on fat metabolism and aging.


Subject(s)
CLOCK Proteins/metabolism , Circadian Clocks , Drosophila Proteins/metabolism , Lipid Metabolism , Longevity , Animals , CLOCK Proteins/genetics , Caloric Restriction , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Gene Expression , Gene Expression Regulation , Male , Signal Transduction
12.
PLoS One ; 8(3): e58773, 2013.
Article in English | MEDLINE | ID: mdl-23554923

ABSTRACT

The bacterial pathogen Bartonella quintana is passed between humans by body lice. B. quintana has adapted to both the human host and body louse vector niches, producing persistent infection with high titer bacterial loads in both the host (up to 10(5) colony-forming units [CFU]/ml) and vector (more than 10(8) CFU/ml). Using a novel custom microarray platform, we analyzed bacterial transcription at temperatures corresponding to the host (37°C) and vector (28°C), to probe for temperature-specific and growth phase-specific transcriptomes. We observed that transcription of 7% (93 genes) of the B. quintana genome is modified in response to change in growth phase, and that 5% (68 genes) of the genome is temperature-responsive. Among these transcriptional changes in response to temperature shift and growth phase was the induction of known B. quintana virulence genes and several previously unannotated genes. Hemin binding proteins, secretion systems, response regulators, and genes for invasion and cell attachment were prominent among the differentially-regulated B. quintana genes. This study represents the first analysis of global transcriptional responses by B. quintana. In addition, the in vivo experiments provide novel insight into the B. quintana transcriptional program within the body louse environment. These data and approaches will facilitate study of the adaptation mechanisms employed by Bartonella during the transition between human host and arthropod vector.


Subject(s)
Bartonella quintana/genetics , Temperature , Transcriptome , Animals , Arthropod Vectors/microbiology , Bartonella quintana/growth & development , Bartonella quintana/pathogenicity , Base Sequence , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Humans , Molecular Sequence Annotation , Nucleotide Motifs , Promoter Regions, Genetic , Transcription, Genetic , Virulence/genetics
13.
Nucleic Acids Res ; 41(12): 5991-6004, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23625967

ABSTRACT

The transcription factor forkhead box P2 (FOXP2) is believed to be important in the evolution of human speech. A mutation in its DNA-binding domain causes severe speech impairment. Humans have acquired two coding changes relative to the conserved mammalian sequence. Despite intense interest in FOXP2, it has remained an open question whether the human protein's DNA-binding specificity and chromatin localization are conserved. Previous in vitro and ChIP-chip studies have provided conflicting consensus sequences for the FOXP2-binding site. Using MITOMI 2.0 microfluidic affinity assays, we describe the binding site of FOXP2 and its affinity profile in base-specific detail for all substitutions of the strongest binding site. We find that human and chimp FOXP2 have similar binding sites that are distinct from previously suggested consensus binding sites. Additionally, through analysis of FOXP2 ChIP-seq data from cultured neurons, we find strong overrepresentation of a motif that matches our in vitro results and identifies a set of genes with FOXP2 binding sites. The FOXP2-binding sites tend to be conserved, yet we identified 38 instances of evolutionarily novel sites in humans. Combined, these data present a comprehensive portrait of FOXP2's-binding properties and imply that although its sequence specificity has been conserved, some of its genomic binding sites are newly evolved.


Subject(s)
DNA/metabolism , Forkhead Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Conserved Sequence , DNA/chemistry , Evolution, Molecular , Forkhead Transcription Factors/genetics , Humans , Microfluidic Analytical Techniques , Mutation , Nucleotide Motifs , Pan troglodytes/genetics , Sequence Analysis, DNA
14.
PLoS One ; 7(10): e48126, 2012.
Article in English | MEDLINE | ID: mdl-23144737

ABSTRACT

Type I interferons (T1IFNs) are among the earliest cytokines produced during infections due to their direct regulation by innate immune signaling pathways. Reports have suggested that T1IFNs are produced during malaria infection, but little is known about the in vivo cellular origins of T1IFNs or their role in protection. We have found that in addition to plasmacytoid dendritic cells, splenic red pulp macrophages (RPMs) can generate significant quantities of T1IFNs in response to P. chabaudi infection in a TLR9-, MYD88-, and IRF7-dependent manner. Furthermore, T1IFNs regulate expression of interferon-stimulated genes redundantly with Interferon-gamma (IFNG), resulting in redundancy for resistance to experimental malaria infection. Despite their role in sensing and promoting immune responses to infection, we observe that RPMs are dispensable for control of parasitemia. Our results reveal that RPMs are early sentinels of malaria infection, but that effector mechanisms previously attributed to RPMs are not essential for control.


Subject(s)
Interferon Type I/immunology , Macrophages/immunology , Malaria/immunology , Plasmodium chabaudi/immunology , Spleen/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Macrophages/metabolism , Malaria/metabolism , Malaria/parasitology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Oligonucleotide Array Sequence Analysis , Plasmodium chabaudi/physiology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/metabolism , Time Factors , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Transcriptome/genetics , Transcriptome/immunology
15.
Proc Natl Acad Sci U S A ; 109(45): E3084-93, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23054834

ABSTRACT

A quantitative understanding of how transcription factors interact with genomic target sites is crucial for reconstructing transcriptional networks in vivo. Here, we use Hac1, a well-characterized basic leucine zipper (bZIP) transcription factor involved in the unfolded protein response (UPR) as a model to investigate interactions between bZIP transcription factors and their target sites. During the UPR, the accumulation of unfolded proteins leads to unconventional splicing and subsequent translation of HAC1 mRNA, followed by transcription of UPR target genes. Initial candidate-based approaches identified a canonical cis-acting unfolded protein response element (UPRE-1) within target gene promoters; however, subsequent studies identified a large set of Hac1 target genes lacking this UPRE-1 and containing a different motif (UPRE-2). Using a combination of unbiased and directed microfluidic DNA binding assays, we established that Hac1 binds in two distinct modes: (i) to short (6-7 bp) UPRE-2-like motifs and (ii) to significantly longer (11-13 bp) extended UPRE-1-like motifs. Using a genetic screen, we demonstrate that a region of extended homology N-terminal to the basic DNA binding domain is required for this dual site recognition. These results establish Hac1 as the first bZIP transcription factor known to adopt more than one binding mode and unify previously conflicting and discrepant observations of Hac1 function into a cohesive model of UPR target gene activation. Our results also suggest that even structurally simple transcription factors can recognize multiple divergent target sites of very different lengths, potentially enriching their downstream target repertoire.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , DNA/metabolism , Microfluidic Analytical Techniques/methods , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Alternative Splicing/genetics , Base Pairing/genetics , Base Sequence , Basic-Leucine Zipper Transcription Factors/chemistry , Binding Sites , Genome/genetics , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Nucleotide Motifs/genetics , Nucleotides/metabolism , Position-Specific Scoring Matrices , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/chemistry , Response Elements/genetics , Saccharomyces cerevisiae Proteins/chemistry , Sequence Homology, Amino Acid , Unfolded Protein Response/genetics
16.
J Neurophysiol ; 96(5): 2593-600, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16855107

ABSTRACT

The thalamic reticular nucleus is strategically located in the axonal pathways between thalamus and cortex, and reticular cells exert strong, topographic inhibition on thalamic relay cells. Although evidence exists that reticular neurons are interconnected through conventional and electrical synapses, the spatial extent and relative strength of these synapses are unclear. To address these issues, we used uncaging of glutamate by laser-scanning photostimulation to provide precisely localized and consistent activation of reticular cell bodies and dendrites in an in vitro slice preparation from the rat as a means to study reticulo-reticular connections. Among the 47 recorded reticular neurons, 29 (62%) received GABAergic axodendritic input from an area immediately surrounding each of the recorded cell bodies, and 8 (17%) responded with depolarizing spikelets, suggesting inputs through electrical synapses. We also found that TTX completely blocked all evoked IPSCs, implying that any dendrodendritic synapses between reticular cells either are relatively weak, have no nearby glutamatergic receptors, or are dependent on back-propagation of action potentials. Finally, we showed that the GABAergic connections between reticular cells are weaker than those from reticular cells to relay cells. Our results suggest that the GABAergic axodendritic synapse is the dominant form of reticulo-reticular connectivity, and because they are much weaker than the reticulo-relay cell synapses, their functional purpose may be to regulate the spatial extent of the reticular inhibition on relay cells.


Subject(s)
Interneurons/physiology , Reticular Formation/physiology , Thalamic Nuclei/physiology , Animals , Axons/physiology , Brain Mapping , Data Interpretation, Statistical , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Gap Junctions/drug effects , Gap Junctions/physiology , Glutamic Acid/metabolism , In Vitro Techniques , Lasers , Neural Pathways/physiology , Photic Stimulation , Pyridazines/pharmacology , Rats , Reticular Formation/cytology , Synapses/physiology , Tetrodotoxin/pharmacology , Thalamic Nuclei/cytology , gamma-Aminobutyric Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...