Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167178, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636614

ABSTRACT

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by haploinsufficiency of transcription factor 4 (TCF4). In this work, we focused on the cerebral cortex and investigated in detail the progenitor cell dynamics and the outcome of neurogenesis in a PTHS mouse model. Labeling and quantification of progenitors and newly generated neurons at various time points during embryonic development revealed alterations affecting the dynamic of cortical progenitors since the earliest stages of cortex formation in PTHS mice. Consequently, establishment of neuronal populations and layering of the cortex were found to be altered in heterozygotes subjects at birth. Interestingly, defective layering process of pyramidal neurons was partially rescued by reintroducing TCF4 expression using focal in utero electroporation in the cerebral cortex. Coincidentally with a defective dorsal neurogenesis, we found that ventral generation of interneurons was also defective in this model, which may lead to an excitation/inhibition imbalance in PTHS. Overall, sex-dependent differences were detected with more marked effects evidenced in males compared with females. All of this contributes to expand our understanding of PTHS, paralleling the advances of research in autism spectrum disorder and further validating the PTHS mouse model as an important tool to advance preclinical studies.


Subject(s)
Cerebral Cortex , Disease Models, Animal , Hyperventilation , Intellectual Disability , Neurogenesis , Transcription Factor 4 , Animals , Transcription Factor 4/metabolism , Transcription Factor 4/genetics , Female , Male , Mice , Hyperventilation/metabolism , Hyperventilation/genetics , Hyperventilation/pathology , Intellectual Disability/genetics , Intellectual Disability/pathology , Intellectual Disability/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Facies , Sex Characteristics , Interneurons/metabolism , Interneurons/pathology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Haploinsufficiency
2.
Semin Cell Dev Biol ; 144: 67-76, 2023 07 30.
Article in English | MEDLINE | ID: mdl-36115764

ABSTRACT

The use of antidepressants during pregnancy benefits the mother's well-being, but the effects of such substances on neurodevelopment remain poorly understood. Moreover, the consequences of early exposure to antidepressants may not be immediately apparent at birth. In utero exposure to selective serotonin reuptake inhibitors (SSRIs) has been related to developmental abnormalities, including a reduced white matter volume. Several reports have observed an increased incidence of autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) after prenatal exposure to SSRIs such as sertraline, the most widely prescribed SSRI. The advent of human-induced pluripotent stem cell (hiPSC) methods and assays now offers appropriate tools to test the consequences of such compounds for neurodevelopment in vitro. In particular, hiPSCs can be used to generate cerebral organoids - self-organized structures that recapitulate the morphology and complex physiology of the developing human brain, overcoming the limitations found in 2D cell culture and experimental animal models for testing drug efficacy and side effects. For example, single-cell RNA sequencing (scRNA-seq) and electrophysiological measurements on organoids can be used to evaluate the impact of antidepressants on the transcriptome and neuronal activity signatures in developing neurons. While the analysis of large-scale transcriptomic data depends on dimensionality reduction methods, electrophysiological recordings rely on temporal data series to discriminate statistical characteristics of neuronal activity, allowing for the rigorous analysis of the effects of antidepressants and other molecules that affect the developing nervous system, especially when applied in combination with relevant human cellular models such as brain organoids.


Subject(s)
Autism Spectrum Disorder , Selective Serotonin Reuptake Inhibitors , Pregnancy , Female , Infant, Newborn , Animals , Humans , Selective Serotonin Reuptake Inhibitors/pharmacology , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/epidemiology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain , Organoids
3.
PLoS Biol ; 20(11): e3001845, 2022 11.
Article in English | MEDLINE | ID: mdl-36327326

ABSTRACT

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), which was rapidly declared a pandemic by the World Health Organization (WHO). Early clinical symptomatology focused mainly on respiratory illnesses. However, a variety of neurological manifestations in both adults and newborns are now well-documented. To experimentally determine whether SARS-CoV-2 could replicate in and affect human brain cells, we infected iPSC-derived human brain organoids. Here, we show that SARS-CoV-2 can productively replicate and promote death of neural cells, including cortical neurons. This phenotype was accompanied by loss of excitatory synapses in neurons. Notably, we found that the U.S. Food and Drug Administration (FDA)-approved antiviral Sofosbuvir was able to inhibit SARS-CoV-2 replication and rescued these neuronal alterations in infected brain organoids. Given the urgent need for readily available antivirals, these results provide a cellular basis supporting repurposed antivirals as a strategic treatment to alleviate neurocytological defects that may underlie COVID-19- related neurological symptoms.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Infant, Newborn , Humans , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Organoids , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Brain , Cell Death , Synapses
4.
Nat Commun ; 13(1): 2387, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35501322

ABSTRACT

Transcription Factor 4 (TCF4) has been associated with autism, schizophrenia, and other neuropsychiatric disorders. However, how pathological TCF4 mutations affect the human neural tissue is poorly understood. Here, we derive neural progenitor cells, neurons, and brain organoids from skin fibroblasts obtained from children with Pitt-Hopkins Syndrome carrying clinically relevant mutations in TCF4. We show that neural progenitors bearing these mutations have reduced proliferation and impaired capacity to differentiate into neurons. We identify a mechanism through which TCF4 loss-of-function leads to decreased Wnt signaling and then to diminished expression of SOX genes, culminating in reduced progenitor proliferation in vitro. Moreover, we show reduced cortical neuron content and impaired electrical activity in the patient-derived organoids, phenotypes that were rescued after correction of TCF4 expression or by pharmacological modulation of Wnt signaling. This work delineates pathological mechanisms in neural cells harboring TCF4 mutations and provides a potential target for therapeutic strategies for genetic disorders associated with this gene.


Subject(s)
Intellectual Disability , Neurons , Cell Proliferation/genetics , Child , Humans , Hyperventilation/metabolism , Intellectual Disability/genetics , Neurons/metabolism , Transcription Factor 4/genetics , Transcription Factor 4/metabolism
5.
J Allergy Clin Immunol ; 149(6): 2171-2176.e3, 2022 06.
Article in English | MEDLINE | ID: mdl-35063500

ABSTRACT

BACKGROUND: Hoffman syndrome is a syndromic, inborn error of immunity due to autosomal-dominant mutations in TOP2B, an essential gene required to alleviate topological stress during DNA replication and gene transcription. Although mutations identified in patients lead to a block in B-cell development and the absence of circulating B cells, an effect on natural killer (NK) cells was not previously examined. OBJECTIVE: We sought to determine whether disease-associated mutations in TOP2B impact NK-cell development and function. METHODS: Using a knockin murine model and patient-derived induced pluripotent stem cells (iPSCs), we investigated NK-cell development in mouse bone marrow and spleen, and performed immunophenotyping by flow cytometry, gene expression, and functional assessment of cytotoxic activity in murine NK cells, and human IPSC-derived NK cells. RESULTS: Mature NK cells were reduced in the periphery of TOP2B knockin mice consistent with patient reports, with reduced cytotoxicity toward target cell lines. IPSCs were successfully derived from patients with Hoffman syndrome, but under optimal conditions showed reduced cytotoxicity compared with iPSC-derived NK cells from healthy controls. CONCLUSIONS: Hoffman syndrome-associated mutations in TOP2B impact NK-cell development and function in murine and human models.


Subject(s)
Induced Pluripotent Stem Cells , Killer Cells, Natural , Animals , Cell Line , Craniofacial Abnormalities , Humans , Induced Pluripotent Stem Cells/metabolism , Limb Deformities, Congenital , Mice , Mutation , Primary Immunodeficiency Diseases , Urogenital Abnormalities
6.
J Vis Exp ; (170)2021 04 11.
Article in English | MEDLINE | ID: mdl-33900298

ABSTRACT

Animals rely on chemical communication to convey and perceive relevant environmental information, ranging from assessment of food quality to detection of available mating partners or threats. In mice, this task is executed primarily by the olfactory system and its underlying subsystems, including the main and accessory olfactory systems. Both have peripheral organs populated by sensory neurons expressing G-protein coupled receptors able to bind chemical cues that reach the nasal cavity. Even though the molecular characteristics of these receptors is well understood, little is known about their cognate specific ligands. The method described here combines in situ hybridization detection of olfactory or vomeronasal receptors with immunodetection of phosphorylated ribosomal protein S6 (pS6) - a marker of neuronal activation. This protocol was devised to identify neurons activated after a single event of exposure to purified or complex chemical stimuli detected by the olfactory organs. Importantly, this technique allows the investigation of neurons triggered in biologically relevant contexts. Ideally, this method should be used to probe the molecular biology of the olfactory system and to study olfactory behaviors.


Subject(s)
Olfactory Receptor Neurons/physiology , Animals , In Situ Hybridization , Male , Mice, Inbred C57BL , Ribosomal Protein S6 , Smell/physiology , Staining and Labeling
7.
Transl Psychiatry ; 11(1): 19, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33414364

ABSTRACT

The human transcription factor 4 gene (TCF4) encodes a helix-loop-helix transcription factor widely expressed throughout the body and during neural development. Mutations in TCF4 cause a devastating autism spectrum disorder known as Pitt-Hopkins syndrome, characterized by a range of aberrant phenotypes including severe intellectual disability, absence of speech, delayed cognitive and motor development, and dysmorphic features. Moreover, polymorphisms in TCF4 have been associated with schizophrenia and other psychiatric and neurological conditions. Details about how TCF4 genetic variants are linked to these diseases and the role of TCF4 during neural development are only now beginning to emerge. Here, we provide a comprehensive review of the functions of TCF4 and its protein products at both the cellular and organismic levels, as well as a description of pathophysiological mechanisms associated with this gene.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Autism Spectrum Disorder/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Facies , Humans , Hyperventilation , Intellectual Disability/genetics , Transcription Factor 4/genetics
8.
STAR Protoc ; 1(3): 100153, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377047

ABSTRACT

This protocol combines fluorescent in situ hybridization and immunostaining to simultaneously detect, in histological sections from the same animal, subpopulations of neurons activated after two episodes of sensory stimulation. It allows the identification of groups of cells singly activated by either stimulus or co-activated by both stimuli. Our method results in nuclear staining for c-Fos mRNA and c-Fos protein, allowing better spatial and temporal resolution than previously published protocols, although it requires quick brain fixation. For complete details on the use and execution of this protocol, please refer to Carvalho et al. (2015, 2020).


Subject(s)
Neurons/cytology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Immunohistochemistry , Mice, Inbred C57BL , RNA/metabolism , Staining and Labeling , Time Factors
9.
Sci Rep ; 10(1): 19943, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203885

ABSTRACT

Behaviors are shaped by hormones, which may act either by changing brain circuits or by modifying sensory detection of relevant cues. Pup-directed behaviors have been previously shown to change via action of hormones at the brain level. Here, we investigated hormonal control of pup-induced activity in the vomeronasal organ, an olfactory sensory structure involved in the detection of non-volatile chemosignals. Vomeronasal activity decreases as males switch from a pup-aggressive state to a non-aggressive parenting state, after they socially contact a female. RNA sequencing, qPCR, and in situ hybridization were used to identify expression, in the vomeronasal sensory epithelium, of candidate GPCR hormone receptors chosen by in silico analyses and educated guesses. After identifying that oxytocin and vasopressin receptors are expressed in the vomeronasal organ, we injected the corresponding hormones in mice and showed that oxytocin administration reduced both pup-induced vomeronasal activity and aggressive behavior. Conversely, injection of an oxytocin receptor antagonist in female-primed male animals, which normally exhibit reduced vomeronasal activity, significantly increased the number of active vomeronasal neurons. These data link oxytocin to the modulation of olfactory sensory activity, providing a possible mechanism for changes in male behavior after social experience with females.


Subject(s)
Aggression/physiology , Biomarkers/analysis , Oxytocics/pharmacology , Oxytocin/pharmacology , Receptors, Oxytocin/metabolism , Vomeronasal Organ/physiology , Aggression/drug effects , Animals , Animals, Newborn , Female , Male , Mice , Oxytocics/administration & dosage , Oxytocin/administration & dosage , RNA-Seq , Vomeronasal Organ/drug effects
10.
Cell Rep ; 32(8): 108061, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32846119

ABSTRACT

The internal representation of sensory information via coherent activation of specific pathways in the nervous system is key to appropriate behavioral responses. Little is known about how chemical stimuli that elicit instinctive behaviors lead to organized patterns of activity in the hypothalamus. Here, we study how a wide range of chemosignals form a discernible map of olfactory information in the ventromedial nucleus of the hypothalamus (VMH) and show that different stimuli entail distinct active neural ensembles. Importantly, we demonstrate that this map depends on functional inputs from the vomeronasal organ. We present evidence that the spatial locations of active VMH ensembles are correlated with activation of distinct vomeronasal receptors and that disjunct VMH ensembles exhibit differential projection patterns. Moreover, active ensembles with distinct spatial locations are not necessarily associated with different behavior categories, such as defensive or social, calling for a revision of the currently accepted model of VMH organization.


Subject(s)
Hypothalamus/physiology , Olfactory Bulb/physiology , Animals , Humans , Mice
11.
Neuron ; 107(1): 8-11, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32645309

ABSTRACT

C1ql3 protein and its receptor Bai3 are involved in synaptic organization and function. In this issue of Neuron, Wang et al. (2020) report that both are essential for synaptic function between the anterior olfactory nucleus and the olfactory bulb and for the generation, but not recall, of associative olfactory memories determining food preference in mice.


Subject(s)
Food Preferences , Synapses , Animals , Memory , Mice , Olfactory Bulb , Smell
12.
DNA Res ; 26(4): 365-378, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31321403

ABSTRACT

Very little is known about long non-coding RNAs (lncRNAs) in the mammalian olfactory sensory epithelia. Deciphering the non-coding transcriptome in olfaction is relevant because these RNAs have been shown to play a role in chromatin modification and nuclear architecture reorganization, processes that accompany olfactory differentiation and olfactory receptor gene choice, one of the most poorly understood gene regulatory processes in mammals. In this study, we used a combination of in silico and ex vivo approaches to uncover a comprehensive catalogue of olfactory lncRNAs and to investigate their expression in the mouse olfactory organs. Initially, we used a novel machine-learning lncRNA classifier to discover hundreds of annotated and unannotated lncRNAs, some of which were predicted to be preferentially expressed in the main olfactory epithelium and the vomeronasal organ, the most important olfactory structures in the mouse. Moreover, we used whole-tissue and single-cell RNA sequencing data to discover lncRNAs expressed in mature sensory neurons of the main epithelium. Candidate lncRNAs were further validated by in situ hybridization and RT-PCR, leading to the identification of lncRNAs found throughout the olfactory epithelia, as well as others exquisitely expressed in subsets of mature olfactory neurons or progenitor cells.


Subject(s)
Machine Learning , Olfactory Receptor Neurons/metabolism , RNA, Long Noncoding/genetics , Transcriptome , Vomeronasal Organ/metabolism , Animals , Female , Male , Mice , RNA, Long Noncoding/metabolism
13.
Methods Mol Biol ; 1820: 289-388, 2018.
Article in English | MEDLINE | ID: mdl-29884953

ABSTRACT

Olfaction is a fundamental sense in most animal species. In mammals, the olfactory system comprises several subpopulations of sensory neurons located throughout the nasal cavity, which detect a variety of chemostimuli, including odorants, intraspecies and interspecies chemical communication cues. Some of these compounds are important for regulating innate and learned behaviors, and endocrine changes in response to other animals in the environment. With a particular focus on laboratory rodent species, this chapter provides a comprehensive description of the most important behavioral assays used for studying the olfactory system, and is meant to be a practical guide for those who study olfaction-mediated behaviors or who have an interest in deciphering the molecular, cellular, or neural mechanisms through which the sense of smell controls the generation of adaptive behavioral outputs.


Subject(s)
Adaptation, Psychological/physiology , Behavior, Animal/physiology , Odorants , Olfactory Perception/physiology , Smell/physiology , Animals , Mice , Rats
14.
Neuron ; 95(4): 734-737, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28817796

ABSTRACT

Territorial male mice are aggressive toward intruding males, but socially bonded males are not. Through manipulation of activity in a subset of neurons in the ventromedial hypothalamus, Yang et al. (2017) report that social and physiological factors non-linearly interact to control male aggression.


Subject(s)
Aggression/physiology , Behavior, Animal/physiology , Brain/physiology , Animals , Humans
15.
Elife ; 62017 04 25.
Article in English | MEDLINE | ID: mdl-28438259

ABSTRACT

The mouse olfactory sensory neuron (OSN) repertoire is composed of 10 million cells and each expresses one olfactory receptor (OR) gene from a pool of over 1000. Thus, the nose is sub-stratified into more than a thousand OSN subtypes. Here, we employ and validate an RNA-sequencing-based method to quantify the abundance of all OSN subtypes in parallel, and investigate the genetic and environmental factors that contribute to neuronal diversity. We find that the OSN subtype distribution is stereotyped in genetically identical mice, but varies extensively between different strains. Further, we identify cis-acting genetic variation as the greatest component influencing OSN composition and demonstrate independence from OR function. However, we show that olfactory stimulation with particular odorants results in modulation of dozens of OSN subtypes in a subtle but reproducible, specific and time-dependent manner. Together, these mechanisms generate a highly individualized olfactory sensory system by promoting neuronal diversity.


Subject(s)
Genetic Variation , Olfactory Pathways/physiology , Olfactory Receptor Neurons/classification , Receptors, Odorant/genetics , Animals , Gene Expression Profiling , Mice , Olfactory Receptor Neurons/physiology , Sequence Analysis, RNA
16.
Sci Rep ; 6: 25507, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27145700

ABSTRACT

The sensory neurons in the olfactory epithelium (OSNs) are equipped with a large repertoire of olfactory receptors and the associated signal transduction machinery. In addition to the canonical OSNs, which express odorant receptors (ORs), the epithelium contains specialized subpopulations of sensory neurons that can detect specific information from environmental cues and relay it to relevant neuronal circuitries. Here we describe a subpopulation of mature OSNs in the main olfactory epithelium (MOE) which expresses CD36, a multifunctional receptor involved in a series of biological processes, including sensory perception of lipid ligands. The Cd36 expressing neurons coexpress markers of mature OSNs and are dispersed throughout the MOE. Unlike several ORs analyzed in our study, we found frequent coexpression of the OR Olfr287 in these neurons, suggesting that only a specific set of ORs may be coexpressed with CD36 in OSNs. We also show that CD36 is expressed in the cilia of OSNs, indicating a possible role in odorant detection. CD36-deficient mice display no signs of gross changes in the organization of the olfactory epithelium, but show impaired preference for a lipid mixture odor. Our results show that CD36-expressing neurons represent a distinct population of OSNs, which may have specific functions in olfaction.


Subject(s)
CD36 Antigens/genetics , GTP-Binding Protein alpha Subunits/genetics , Olfactory Marker Protein/genetics , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/genetics , Animals , CD36 Antigens/deficiency , Cilia/drug effects , Cilia/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation , Lipids/pharmacology , Male , Mice , Mice, Knockout , Odorants/analysis , Olfactory Marker Protein/metabolism , Olfactory Mucosa/cytology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/drug effects , Pheromones/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Odorant/metabolism , Smell/physiology
17.
BMC Biol ; 14: 12, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26878847

ABSTRACT

BACKGROUND: Olfaction is a fundamental sense through which most animals perceive the external world. The olfactory system detects odors via specialized sensory organs such as the main olfactory epithelium and the vomeronasal organ. Sensory neurons in these organs use G-protein coupled receptors to detect chemosensory stimuli. The odorant receptor (OR) family is expressed in sensory neurons of the main olfactory epithelium, while the adult vomeronasal organ is thought to express other types of receptors. RESULTS: Here, we describe Olfr692, a member of the OR gene family identified by next-generation RNA sequencing, which is highly upregulated and non-canonically expressed in the vomeronasal organ. We show that neurons expressing this gene are activated by odors emanating from pups. Surprisingly, activity in Olfr692-positive cells is sexually dimorphic, being very low in females. Our results also show that juvenile odors activate a large number of Olfr692 vomeronasal neurons in virgin males, which is correlated with the display of infanticide behavior. . In contrast, activity substantially decreases in parenting males (fathers), where infanticidal aggressive behavior is not frequently observed. CONCLUSIONS: Our results describe, for the first time, a sensory neural population with a specific molecular identity involved in the detection of pup odors. Moreover, it is one of the first reports of a group of sensory neurons the activity of which is sexually dimorphic and depends on social status. Our data suggest that the Olfr692 population is involved in mediating pup-oriented behaviors in mice.


Subject(s)
Odorants , Receptors, Odorant/genetics , Sensory Receptor Cells/metabolism , Smell , Vomeronasal Organ/cytology , Aggression , Animals , Animals, Newborn , Behavior, Animal , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Odorants/analysis , Receptors, Odorant/analysis , Sex Characteristics , Vomeronasal Organ/physiology
18.
Front Neurosci ; 9: 283, 2015.
Article in English | MEDLINE | ID: mdl-26321906

ABSTRACT

The nervous system is organized to detect, internally represent and process sensory information to generate appropriate behaviors. Despite the crucial importance of odors that elicit instinctive behaviors, such as pheromones and kairomones, their neural representation remains little characterized in the mammalian brain. Here we used expression of the immediate early gene product c-Fos as a marker of neuronal activity to find that a wide range of pheromones and kairomones produces activation in the medial nucleus of the amygdala, a brain area anatomically connected with the olfactory sensory organs. We see that activity in this nucleus depends on vomeronasal organ input, and that distinct vomeronasal stimuli activate a dispersed ensemble of cells, without any apparent spatial segregation. This activity pattern does not reflect the chemical category of the stimuli, their valence or the induced behaviors. These findings will help build a complete understanding of how odor information is processed in the brain to generate instinctive behaviors.

19.
BMC Genomics ; 11: 462, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20691070

ABSTRACT

BACKGROUND: To date, oil-rich plants are the main source of biodiesel products. Because concerns have been voiced about the impact of oil-crop cultivation on the price of food commodities, the interest in oil plants not used for food production and amenable to cultivation on non-agricultural land has soared. As a non-food, drought-resistant and oil-rich crop, Jatropha curcas L. fulfils many of the requirements for biofuel production. RESULTS: We have generated 13,249 expressed sequence tags (ESTs) from developing and germinating Jatropha seeds. This strategy allowed us to detect most known genes related to lipid synthesis and degradation. We have also identified ESTs coding for proteins that may be involved in the toxicity of Jatropha seeds. Another unexpected finding is the high number of ESTs containing transposable element-related sequences in the developing seed library (800) when contrasted with those found in the germinating seed library (80). CONCLUSIONS: The sequences generated in this work represent a considerable increase in the number of sequences deposited in public databases. These results can be used to produce genetically improved varieties of Jatropha with increased oil yields, different oil compositions and better agronomic characteristics.


Subject(s)
Jatropha/genetics , Plant Oils/analysis , DNA Transposable Elements , Databases, Nucleic Acid , Gene Expression Profiling , Germination , Jatropha/chemistry , Jatropha/metabolism , Plant Oils/metabolism , Seeds/chemistry , Seeds/genetics , Seeds/metabolism , Transcription, Genetic
20.
Cell ; 141(4): 692-703, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20478258

ABSTRACT

Potential predators emit uncharacterized chemosignals that warn receiving species of danger. Neurons that sense these stimuli remain unknown. Here we show that detection and processing of fear-evoking odors emitted from cat, rat, and snake require the function of sensory neurons in the vomeronasal organ. To investigate the molecular nature of the sensory cues emitted by predators, we isolated the salient ligands from two species using a combination of innate behavioral assays in naive receiving animals, calcium imaging, and c-Fos induction. Surprisingly, the defensive behavior-promoting activity released by other animals is encoded by species-specific ligands belonging to the major urinary protein (Mup) family, homologs of aggression-promoting mouse pheromones. We show that recombinant Mup proteins are sufficient to activate sensory neurons and initiate defensive behavior similarly to native odors. This co-option of existing sensory mechanisms provides a molecular solution to the difficult problem of evolving a variety of species-specific molecular detectors.


Subject(s)
Behavior, Animal , Pheromones/metabolism , Vomeronasal Organ/metabolism , Animals , Cats , Female , Male , Mice , Mice, Inbred C57BL , Odorants , Predatory Behavior , Proteins/metabolism , Rats , Recombinant Proteins/metabolism , Snakes , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...