480 research outputs found

    Transcript expression of vesicular glutamate transporters in lumbar dorsal root ganglia and the spinal cord of mice – Effects of peripheral axotomy or hindpaw inflammation

    Get PDF
    Using specific riboprobes, we characterized the expression of vesicular glutamate transporter (VGLUT)1–VGLUT3 transcripts in lumbar 4–5 (L4–5) dorsal root ganglions (DRGs) and the thoracolumbar to lumbosacral spinal cord in male BALB/c mice after a 1- or 3-day hindpaw inflammation, or a 7-day sciatic nerve axotomy. Sham animals were also included. In sham and contralateral L4–5 DRGs of injured mice, VGLUT1-, VGLUT2- and VGLUT3 mRNAs were expressed in ∼45%, ∼69% or ∼17% of neuron profiles (NPs), respectively. VGLUT1 was expressed in large and medium-sized NPs, VGLUT2 in NPs of all sizes, and VGLUT3 in small and medium-sized NPs. In the spinal cord, VGLUT1 was restricted to a number of NPs at thoracolumbar and lumbar segments, in what appears to be the dorsal nucleus of Clarke, and in mid laminae III–IV. In contrast, VGLUT2 was present in numerous NPs at all analyzed spinal segments, except the lateral aspects of the ventral horns, especially at the lumbar enlargement, where it was virtually absent. VGLUT3 was detected in a discrete number of NPs in laminae III–IV of the dorsal horn. Axotomy resulted in a moderate decrease in the number of DRG NPs expressing VGLUT3, whereas VGLUT1 and VGLUT2 were unaffected. Likewise, the percentage of NPs expressing VGLUT transcripts remained unaltered after hindpaw inflammation, both in DRGs and the spinal cord. Altogether, these results confirm previous descriptions on VGLUTs expression in adult mice DRGs, with the exception of VGLUT1, whose protein expression was detected in a lower percentage of mouse DRG NPs. A detailed account on the location of neurons expressing VGLUTs transcripts in the adult mouse spinal cord is also presented. Finally, the lack of change in the number of neurons expressing VGLUT1 and VGLUT2 transcripts after axotomy, as compared to data on protein expression, suggests translational rather than transcriptional regulation of VGLUTs after injury.Fil: Malet, Mariana. Universidad Austral. Facultad de Ciencias Biomédicas; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas; ArgentinaFil: Vieytes, C. A.. Universidad Austral. Facultad de Ciencias Biomédicas; ArgentinaFil: Lundgren, K. H.. University of Cincinnati; Estados UnidosFil: Seal, R. P.. University of Pittsburgh; Estados UnidosFil: Tomasella, María Eugenia. Universidad Austral. Facultad de Ciencias Biomédicas; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas; ArgentinaFil: Seroogy, K. B.. University of Cincinnati; Estados UnidosFil: Hökfelt, T.. Karolinska Huddinge Hospital. Karolinska Institutet; SueciaFil: Gebhart, G. F.. University of Pittsburgh; Estados UnidosFil: Brumovsky, Pablo Rodolfo. Universidad Austral. Facultad de Ciencias Biomédicas; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. University of Pittsburgh; Estados Unido

    Serotoninergic, peptidergic and GABAergic innervation of the ventrolateral and dorsolateral motor nuclei in the cat S1/S2 segments: An immunofluorescence study

    Get PDF
    Indirect single- and double-staining immunofluorescence techniques were used to study the serotoninergic, peptidergic and GABAergic innervation of the ventrolateral (Onuf's nucleus) and dorsolateral (innervating intrinsic foot sole muscles) nuclei, located in the S1/S2 segments of the cat spinal cord. The relative density of 5-hydroxytryptamine-, thyrotropin-releasing hormone-, substance P- and γ-aminobuytric acid-immunoreactive axonal varicosities was similar in both nuclei. The highest relative density was recorded for varicosities immunoreactive to γ-aminobutyric acid, while those immunoreactive to 5-hydroxytryptamine or thyrotropin-releasing hormone yielded the lowest values. The density of enkephalin-immunoreactive varicosities was higher in the ventrolateral than in the dorsolateral nucleus. Calcitonin gene-related peptide-like immunoreactivity could be seen in neurons of the ventrolateral and dorsolateral nuclei. Occasionally, calcitonin gene-related peptide-immunoreactive axonal fibers were also encountered in these nuclei. Virtually all thyrotropin-releasing hormone-immunoreactive varicosities in the ventrolateral and dorsolateral nuclei also contained 5-hydroxytryptamine-like immunoreactivity, while a somewhat smaller number of them were co-localized with substance P. About 5–10% of the 5-hydroxytryptamine-immunoreactive varicosities were devoid of peptide-like immunoreactivity, and the number of 5-hydroxytryptamine-immunoreactive varicosities lacking thyrotropin-releasing hormone-like immunoreactivity was higher in the dorsolateral than in the ventrolateral nucleus. Finally, the free fraction of substance P-immunoreactive varicosities, i.e., those lacking both 5-hydroxytryptamine and thyrotropin-releasing hormone, was about 39% in the ventrolateral and 26% in the dorsolateral nucleus. Spinal cord transection at the lower thoracic level induced a depletion of 5-hydroxytryptamine and thyrotropin-releasing hormone-immunoreactive fibers from the ventrolateral and dorsolateral nuclei, indicating an exclusive supraspinal origin for these fibers. A reduction in substance P-like immunoreactivity following spinal cord transection alone or spinal cord transection combined with unilateral dorsal rhizotomy was also detected in both nuclei, suggesting a dual origin for substance P-immunoreactive fibers, i.e., both supra- and intraspinal. The decrease in number of substance P-immunoreactive fibers was however smaller than expected from the analysis of the fraction of substance P-immunoreactive fibers co-localized with 5-hydroxytryptamine, indicating thus that the experimental lesions may have triggered a sprouting of substance P-immunoreactive axons originating from spinal cord sources. The distribution of γ-aminobutyric acid in the ventrolateral and dorsolateral nuclei was not affected by the different lesion paradigms. It is therefore assumed that these inputs are intrinsic to the spinal cord. Finally, both in the ventrolateral and the dorsolateral nucleus a small but statistically significant increase of axonal fibers immunoreactive to enkephalin was seen in response to the experimental lesions

    Some lumbar sympathetic neurons develop a glutamatergic phenotype after peripheral axotomy with a note on VGLUT2-positive perineuronal baskets

    Get PDF
    Glutamate is the main excitatory neurotransmitter in the nervous system, including in primary afferent neurons. However, to date a glutamatergic phenotype of autonomic neurons has not been described. Therefore, we explored the expression of vesicular glutamate transporter (VGLUT) types 1, 2 and 3 in lumbar sympathetic chain (LSC) and major pelvic ganglion (MPG) of naïve BALB/C mice, as well as after pelvic nerve axotomy (PNA), using immunohistochemistry and in situ hybridization. Colocalization with activating transcription factor-3 (ATF-3), tyrosine hydroxylase (TH), vesicular acetylcholine transporter (VAChT) and calcitonin gene-related peptide was also examined. Sham-PNA, sciatic nerve axotomy (SNA) or naïve mice were included. In naïve mice, VGLUT2-like immunoreactivity (LI) was only detected in fibers and varicosities in LSC and MPG; no ATF-3-immunoreactive (IR) neurons were visible. In contrast, PNA induced upregulation of VGLUT2 protein and transcript, as well as of ATF-3-LI in subpopulations of LSC neurons. Interestingly, VGLUT2-IR LSC neurons coexpressed ATF-3, and often lacked the noradrenergic marker TH. SNA only increased VGLUT2 protein and transcript in scattered LSC neurons. Neither PNA nor SNA upregulated VGLUT2 in MPG neurons. We also found perineuronal baskets immunoreactive either for VGLUT2 or the acetylcholinergic marker VAChT in non-PNA MPGs, usually around TH-IR neurons. VGLUT1-LI was restricted to some varicosities in MPGs, was absent in LSCs, and remained largely unaffected by PNA or SNA. This was confirmed by the lack of expression of VGLUT1 or VGLUT3 mRNAs in LSCs, even after PNA or SNA. Taken together, axotomy of visceral and non-visceral nerves results in a glutamatergic phenotype of some LSC neurons. In addition, we show previously non-described MPG perineuronal glutamatergic baskets.Fil: Brumovsky, Pablo Rodolfo. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Universidad Austral; Argentina. Univeristy of Pittsburgh. School of Medicine; Estados UnidosFil: Seroogy, Kim B.. University of Cincinnati; Estados UnidosFil: Lundgren, Kerstin H.. University of Cincinnati; Estados UnidosFil: Watanabe, Masahiko. Hokkaido University School of Medicine; JapónFil: Hökfelt, Tomas. Karolinska Huddinge Hospital. Karolinska Institutet; SueciaFil: Gebhart, G.F.. Univeristy of Pittsburgh. School of Medicine; Estados Unido

    Responses of substantia gelatinosa neurons to putative neurotransmitters in an in vitro preparation of the adult rat spinal cord

    Get PDF
    Extracellular recordings were performed from neurons of the substantia gelatinosa (SG) in an in vitro preparation obtained from the spinal cord of adult rats. About 40% of neurons were spontaneously active. They could be synaptically influenced by low and high threshold fiber input entering the spinal cord through dorsal and ventral and ventral roots. Repetitive low threshold stimulation led to a transient increase in activity of a number of these neurons, whereas high intensity stimulation predominantly reduced excitability. The majority of non-spontaneously active neurons responded to an increase of stimulus intensity covariantly with an increase in firing rate. The excitatory effect of phoretically administeredl-glutamate as well as synaptically induced and spontaneous activity was reduced or abolished by phoretically administered GABA, glycine or the enkephalin-analogued-Ala2-d-Leu5-enkephalin. The actions of the enkephalin analogue were blocked by phoretically applied naloxone. The findings are consistent with the notion from in vivo investigations of a structurally and functionally heterogeneous population of neurons which display a responsiveness to microtopically applied putative neurotransmitters resembling dorsal horn neurons in deeper layers

    Expression of vesicular glutamate transporters type 1 and 2 in sensory and autonomic neurons innervating the mouse colorectum

    Get PDF
    Vesicular glutamate transporters (VGLUTs) have been extensively studied in various neuronal systems, but their expression in visceral sensory and autonomic neurons remains to be analyzed in detail. Here we studied VGLUTs type 1 and 2 (VGLUT(1) and VGLUT(2) , respectively) in neurons innervating the mouse colorectum. Lumbosacral and thoracolumbar dorsal root ganglion (DRG), lumbar sympathetic chain (LSC), and major pelvic ganglion (MPG) neurons innervating the colorectum of BALB/C mice were retrogradely traced with Fast Blue, dissected, and processed for immunohistochemistry. Tissue from additional naïve mice was included. Previously characterized antibodies against VGLUT(1) , VGLUT(2) , and calcitonin gene-related peptide (CGRP) were used. Riboprobe in situ hybridization, using probes against VGLUT(1) and VGLUT(2) , was also performed. Most colorectal DRG neurons expressed VGLUT(2) and often colocalized with CGRP. A smaller percentage of neurons expressed VGLUT(1) . VGLUT(2) -immunoreactive (IR) neurons in the MPG were rare. Abundant VGLUT(2) -IR nerves were detected in all layers of the colorectum; VGLUT(1) -IR nerves were sparse. A subpopulation of myenteric plexus neurons expressed VGLUT2 protein and mRNA, but VGLUT1 mRNA was undetectable. In conclusion, we show 1) that most colorectal DRG neurons express VGLUT(2) , and to a lesser extent, VGLUT(1) ; 2) abundance of VGLUT2-IR fibers innervating colorectum; and 3) a subpopulation of myenteric plexus neurons expressing VGLUT(2). Altogether, our data suggests a role for VGLUT(2) in colorectal glutamatergic neurotransmission, potentially influencing colorectal sensitivity and motility.Fil: Brumovsky, Pablo Rodolfo. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Univeristy of Pittsburgh. School of Medicine; Estados Unidos. Universidad Austral; ArgentinaFil: Robinson, David R.. Univeristy of Pittsburgh. School of Medicine; Estados Unidos. University of Pittsburgh at Johnstown; Estados Unidos. University of Pittsburgh; Estados UnidosFil: La, Jun Ho. Univeristy of Pittsburgh. School of Medicine; Estados UnidosFil: Seroogy, Kim B.. No especifíca;Fil: Lundgren, Kerstin H.. No especifíca;Fil: Albers, Kathryn M.. Univeristy of Pittsburgh. School of Medicine; Estados UnidosFil: Kiyatkin, Michael E.. Univeristy of Pittsburgh. School of Medicine; Estados UnidosFil: Seal, Rebecca P.. No especifíca;Fil: Edwards, Robert H.. No especifíca;Fil: Watanabe, Masahiko. No especifíca;Fil: Hökfelt, Tomas. Karolinska Huddinge Hospital. Karolinska Institutet; SueciaFil: Gebhart, G. F.. Univeristy of Pittsburgh. School of Medicine; Estados Unido

    Circuit dissection of the role of somatostatin in itch and pain

    Get PDF
    Stimuli that elicit itch are detected by sensory neurons that innervate the skin. This information is processed by the spinal cord; however, the way in which this occurs is still poorly understood. Here we investigated the neuronal pathways for itch neurotransmission, particularly the contribution of the neuropeptide somatostatin. We find that in the periphery, somatostatin is exclusively expressed in Nppb+ neurons, and we demonstrate that Nppb+somatostatin+ cells function as pruriceptors. Employing chemogenetics, pharmacology and cell-specific ablation methods, we demonstrate that somatostatin potentiates itch by inhibiting inhibitory dynorphin neurons, which results in disinhibition of GRPR+ neurons. Furthermore, elimination of somatostatin from primary afferents and/or from spinal interneurons demonstrates differential involvement of the peptide released from these sources in itch and pain. Our results define the neural circuit underlying somatostatin-induced itch and characterize a contrasting antinociceptive role for the peptide

    A Comparative Study of Two Blast-Induced Traumatic Brain Injury Models: Changes in Monoamine and Galanin Systems Following Single and Repeated Exposure

    Get PDF
    Repeated mild blast-induced traumatic brain injury (rmbTBI), caused by recurrent exposure to low levels of explosive blast, is a significant concern for military health systems. However, the pathobiology of rmbTBI is currently poorly understood. Animal models are important tools to identify the molecular changes of rmbTBI, but comparisons across different models can present their own challenges. In this study, we compared two well-established rodent models of mbTBI, the “KI model” and the “USU/WRAIR model.” These two models create different pulse forms, in terms of peak pressure and duration. Following single and double exposures to mild levels of blast, we used in situ hybridization (ISH) to assess changes in mRNA levels of tyrosine hydroxylase (TH), tryptophan hydroxylase (TPH2), and galanin in the locus coeruleus (LC) and dorsal raphe nucleus (DRN). These systems and their transmitters are known to mediate responses to stress and anxiety. We found increased mRNA levels of TH, TPH2 and galanin in the LC and DRN of single-exposed rats relative to sham rats in the KI but not the USU/WRAIR model. Sham mRNA values measured in the USU/WRAIR model were substantially higher than their KI counterparts. Double exposure caused similarly significant increases in mRNA values in the KI model but not the USU/WRAIR model, except TPH2 and galanin levels in the DRN. We detected no cumulative effect of injury in either model at the used inter-injury interval (30 min), and there were no detectable neuropathological changes in any experimental group at 1 day post-injury. The apparent lack of early response to injury as compared to sham in the USU/WRAIR model is likely caused by stressors (e.g., transportation and noise), associated with the experimental execution, that were absent in the KI model. This study is the first to directly compare two established rodent models of rmbTBI, and to highlight the challenges of comparing findings from different animal models. Additional studies are needed to understand the role of stress, dissect the effects of psychological and physical injuries and to identify the window of increased cerebral vulnerability, i.e., the inter-injury interval that results in a cumulative effect following repeated blast exposure

    Distinct transcriptional responses of mouse sensory neurons in models of human chronic pain conditions.

    Get PDF
    Background: Sensory neurons play an essential role in almost all pain conditions, and have recently been classified into distinct subsets on the basis of their transcriptomes. Here we have analysed alterations in dorsal root ganglia (DRG) gene expression using microarrays in mouse models related to human chronic pain. Methods: Six different pain models were studied in male C57BL/6J mice: (1) bone cancer pain using cancer cell injection in the intramedullary space of the femur; (2) neuropathic pain using partial sciatic nerve ligation; (3) osteoarthritis pain using mechanical joint loading; (4) chemotherapy-induced pain with oxaliplatin; (5) chronic muscle pain using hyperalgesic priming; and (6) inflammatory pain using intraplantar complete Freund's adjuvant. Microarray analyses were performed using RNA isolated from dorsal root ganglia and compared to sham/vehicle treated controls. Results: Differentially expressed genes (DEGs) were identified. Known and previously unreported genes were found to be dysregulated in each pain model. The transcriptomic profiles for each model were compared and expression profiles of DEGs within subsets of DRG neuronal populations were analysed to determine whether specific neuronal subsets could be linked to each of the pain models.  Conclusions: Each pain model exhibits a unique set of altered transcripts implying distinct cellular responses to different painful stimuli. No simple direct link between genetically distinct sets of neurons and particular pain models could be discerned

    New Mouse Lines for the Analysis of Neuronal Morphology Using CreER(T)/loxP-Directed Sparse Labeling

    Get PDF
    BACKGROUND: Pharmacologic control of Cre-mediated recombination using tamoxifen-dependent activation of a Cre-estrogen receptor ligand binding domain fusion protein [CreER(T)] is widely used to modify and/or visualize cells in the mouse. METHODS AND FINDINGS: We describe here two new mouse lines, constructed by gene targeting to the Rosa26 locus to facilitate Cre-mediated cell modification. These lines should prove particularly useful in the context of sparse labeling experiments. The R26rtTACreER line provides ubiquitous expression of CreER under transcriptional control by the tetracycline reverse transactivator (rtTA); dual control by doxycycline and tamoxifen provides an extended dynamic range of Cre-mediated recombination activity. The R26IAP line provides high efficiency Cre-mediated activation of human placental alkaline phosphatase (hPLAP), complementing the widely used, but low efficiency, Z/AP line. By crossing with mouse lines that direct cell-type specific CreER expression, the R26IAP line has been used to produce atlases of labeled cholinergic and catecholaminergic neurons in the mouse brain. The R26IAP line has also been used to visualize the full morphologies of retinal dopaminergic amacrine cells, among the largest neurons in the mammalian retina. CONCLUSIONS: The two new mouse lines described here expand the repertoire of genetically engineered mice available for controlled in vivo recombination and cell labeling using the Cre-lox system
    corecore