151
|
Abstract
The transcription factor Runt-related transcription factor 1 (RUNX1) is critical for the earliest steps of hematopoiesis. RUNX1 was originally identified as a gene fusion in acute myeloid leukemia (AML) and thus has garnered heavy attention as a tumor suppressor in hematopoietic malignancies. However, RUNX1 is also strongly expressed in breast epithelia and may be misregulated during tumorigenesis. Here, I discuss our recent work implicating RUNX1 in proliferation control during breast epithelial-acinar morphogenesis. My goal is to place these findings in the context of a handful of other reports, which together argue that RUNX1 could act as a tumor suppressor gene in breast cancer. Testing this hypothesis requires focused in vivo studies, because the major commercial platform for global mRNA expression profiling does not reliably reflect RUNX1 levels. Our in vitro results indicate that hyperproliferation in RUNX1-deficient breast epithelia relies on another family of transcription factors, the Forkhead box O (FOXO) proteins. FOXOs could, therefore, represent a synthetic-lethal target for RUNX1-deficient tumors if the hypothesized link to breast cancer is correct.
Collapse
Affiliation(s)
- Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
152
|
Gardiner NJ. Integrins and the extracellular matrix: Key mediators of development and regeneration of the sensory nervous system. Dev Neurobiol 2011; 71:1054-72. [DOI: 10.1002/dneu.20950] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
153
|
Stein C, Machelska H. Modulation of Peripheral Sensory Neurons by the Immune System: Implications for Pain Therapy. Pharmacol Rev 2011; 63:860-81. [DOI: 10.1124/pr.110.003145] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
154
|
The transcription factor Smad-interacting protein 1 controls pain sensitivity via modulation of DRG neuron excitability. Pain 2011; 152:2384-2398. [DOI: 10.1016/j.pain.2011.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 06/20/2011] [Accepted: 07/11/2011] [Indexed: 11/21/2022]
|
155
|
Restriction of transient receptor potential vanilloid-1 to the peptidergic subset of primary afferent neurons follows its developmental downregulation in nonpeptidergic neurons. J Neurosci 2011; 31:10119-27. [PMID: 21752988 DOI: 10.1523/jneurosci.1299-11.2011] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Primary afferent "pain" fibers (nociceptors) are divided into subclasses based on distinct molecular and anatomical features, and these classes mediate noxious modality-specific contributions to behaviors evoked by painful stimuli. Whether the heat and capsaicin receptor transient receptor potential vanilloid-1 (TRPV1) is expressed heterogeneously across several sensory populations, or is selectively expressed by a unique nociceptor subclass, however, is unclear. Here we used two lines of Trpv1 reporter mice to investigate the primary afferent expression of TRPV1, both during development and in the adult. We demonstrate, using Cre-induced lineage tracing, that during development TRPV1 is transiently expressed in a wide range of dorsal root ganglion neurons, and that its expression is gradually refined, such that TRPV1 transcripts become restricted to a specific subset of peptidergic sensory neurons. Finally, the remarkable sensitivity that is characteristic of these reporter mice revealed an innervation of central and peripheral targets by TRPV1+ primary afferents in the adult that is considerably more extensive than has previously been appreciated.
Collapse
|
156
|
Levanon D, Bernstein Y, Negreanu V, Bone KR, Pozner A, Eilam R, Lotem J, Brenner O, Groner Y. Absence of Runx3 expression in normal gastrointestinal epithelium calls into question its tumour suppressor function. EMBO Mol Med 2011; 3:593-604. [PMID: 21786422 PMCID: PMC3258485 DOI: 10.1002/emmm.201100168] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/07/2011] [Accepted: 07/18/2011] [Indexed: 11/15/2022] Open
Abstract
The Runx3 transcription factor regulates cell fate decisions during embryonic development and in adults. It was previously reported that Runx3 is strongly expressed in embryonic and adult gastrointestinal tract (GIT) epithelium (Ep) and that its loss causes gastric cancer. More than 280 publications have based their research on these findings and concluded that Runx3 is indeed a tumour suppressor (TS). In stark contrast, using various measures, we found that Runx3 expression is undetectable in GIT Ep. Employing a variety of biochemical and genetic techniques, including analysis of Runx3-GFP and R26LacZ/Runx3Cre or R26tdTomato/Runx3Cre reporter strains, we readily detected Runx3 in GIT-embedded leukocytes, dorsal root ganglia, skeletal elements and hair follicles. However, none of these approaches revealed detectable Runx3 levels in GIT Ep. Moreover, our analysis of the original Runx3LacZ/LacZ mice used in the previously reported study failed to reproduce the GIT expression of Runx3. The lack of evidence for Runx3 expression in normal GIT Ep creates a serious challenge to the published data and undermines the notion that Runx3 is a TS involved in cancer pathogenesis.
Collapse
Affiliation(s)
- Ditsa Levanon
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Transcription factor short stature homeobox 2 is required for proper development of tropomyosin-related kinase B-expressing mechanosensory neurons. J Neurosci 2011; 31:6741-9. [PMID: 21543603 DOI: 10.1523/jneurosci.5883-10.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Dorsal root ganglia (DRG) contain somatosensory neurons of diverse sensory modalities. Among these different types of sensory neurons, the molecular mechanisms that regulate the development and specification of touch neurons are the least well understood. We took a candidate approach and searched for transcription factors that are expressed in subsets of DRG neurons, and found that the transcription factor Shox2 (short stature homeobox 2) is expressed in subpopulations of TrkB (tropomyosin-related kinase B)- and Ret-expressing neurons at neonatal stages. Since TrkB is a known marker that is selectively expressed in touch sensory neurons, we decided to examine the function of Shox2 in specifying TrkB-positive DRG neurons. Conditional deletion of Shox2 in neural crest cells (which give rise to all DRG neurons) caused a 60 ∼ 65% reduction in the number of TrkB-expressing neurons. It also resulted in an increase in coexpression of TrkC in Ret-positive sensory neurons. Deletion of Shox2 in differentiating DRG neurons at later time points caused only a moderate reduction in TrkB expression. Overexpression of Shox2 in all neural crest cells resulted in a small increase in the number of TrkB-expressing neurons. Finally, Shox2 deletion also caused reduced touch sensory axonal innervation to layers III/IV of the spinal cord. Together, our findings identify Shox2 as an essential but not sufficient component of the transcription programs required in neural progenitor cells for the proper specification of subsets of TrkB-expressing touch/mechanosensory neurons.
Collapse
|
158
|
König N, Åkesson E, Telorack M, Vasylovska S, Ngamjariyawat A, Sundström E, Oster A, Trolle C, Berens C, Aldskogius H, Seiger Å, Kozlova EN. Forced Runx1 expression in human neural stem/progenitor cells transplanted to the rat dorsal root ganglion cavity results in extensive axonal growth specifically from spinal cord-derived neurospheres. Stem Cells Dev 2011; 20:1847-57. [PMID: 21322790 DOI: 10.1089/scd.2010.0555] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell replacement therapy holds great promise for treating a wide range of human disorders. However, ensuring the predictable differentiation of transplanted stem cells, eliminating their risk of tumor formation, and generating fully functional cells after transplantation remain major challenges in regenerative medicine. Here, we explore the potential of human neural stem/progenitor cells isolated from the embryonic forebrain (hfNSPCs) or the spinal cord (hscNSPCs) to differentiate to projection neurons when transplanted into the dorsal root ganglion cavity of adult recipient rats. To stimulate axonal growth, we transfected hfNSPC- and hscNSPC-derived neurospheres, prior to their transplantation, with a Tet-Off Runx1-overexpressing plasmid to maintain Runx1 expression in vivo after transplantation. Although pronounced cell differentiation was found in the Runx1-expressing transplants from both cell sources, we observed extensive, long-distance growth of axons exclusively from hscNSPC-derived transplants. These axons ultimately reached the dorsal root transitional zone, the boundary separating peripheral and central nervous systems. Our data show that hscNSPCs have the potential to differentiate to projection neurons with long-distance axonal outgrowth and that Runx1 overexpression is a useful approach to induce such outgrowth in specific sources of NSPCs.
Collapse
Affiliation(s)
- Niclas König
- Department of Neuroscience, Neuroanatomy, Uppsala University Biomedical Center, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Abstract
Small fiber sensory neuropathy is a common disorder in which progressive degeneration of small-diameter nociceptors causes decreased sensitivity to thermal stimuli and painful sensations in the extremities. In the majority of patients, the cause of small fiber sensory neuropathy is unknown, and treatment options are limited. Here, we show that Bcl-w (Bcl-2l2) is required for the viability of small fiber nociceptive sensory neurons. Bcl-w(-/-) mice demonstrate an adult-onset progressive decline in thermosensation and a decrease in nociceptor innervation of the epidermis. This denervation occurs without cell body loss, indicating that lack of Bcl-w results in a primary axonopathy. Consistent with this phenotype, we show that Bcl-w, in contrast to the closely related Bcl-2 and Bcl-xL, is enriched in axons of sensory neurons and that Bcl-w prevents the dying back of axons. Bcl-w(-/-) sensory neurons exhibit mitochondrial abnormalities, including alterations in axonal mitochondrial size, axonal mitochondrial membrane potential, and cellular ATP levels. Collectively, these data establish bcl-w(-/-) mice as an animal model of small fiber sensory neuropathy and provide new insight regarding the role of Bcl-w and of mitochondria in preventing axonal degeneration.
Collapse
|
160
|
Franck MCM, Stenqvist A, Li L, Hao J, Usoskin D, Xu X, Wiesenfeld-Hallin Z, Ernfors P. Essential role of Ret for defining non-peptidergic nociceptor phenotypes and functions in the adult mouse. Eur J Neurosci 2011; 33:1385-400. [DOI: 10.1111/j.1460-9568.2011.07634.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
161
|
Vaegter CB, Jansen P, Fjorback AW, Glerup S, Skeldal S, Richner M, Erdmann B, Nyengaard JR, Tessarollo L, Lewin GR, Willnow TE, Chao MV, Nykjaer A. Sortilin associates with Trk receptors to enhance anterograde transport and neurotrophin signaling. Nat Neurosci 2011; 14:54-61. [PMID: 21102451 PMCID: PMC3808973 DOI: 10.1038/nn.2689] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 10/12/2010] [Indexed: 02/05/2023]
Abstract
Binding of target-derived neurotrophins to Trk receptors at nerve terminals is required to stimulate neuronal survival, differentiation, innervation and synaptic plasticity. The distance between the soma and nerve terminal is great, making efficient anterograde Trk transport critical for Trk synaptic translocation and signaling. The mechanism responsible for this trafficking remains poorly understood. Here we show that the sorting receptor sortilin interacts with TrkA, TrkB and TrkC and enables their anterograde axonal transport, thereby enhancing neurotrophin signaling. Cultured DRG neurons lacking sortilin showed blunted MAP kinase signaling and reduced neurite outgrowth upon stimulation with NGF. Moreover, deficiency for sortilin markedly aggravated TrkA, TrkB and TrkC phenotypes present in p75(NTR) knockouts, and resulted in increased embryonic lethality and sympathetic neuropathy in mice heterozygous for TrkA. Our findings demonstrate a role for sortilin as an anterograde trafficking receptor for Trk and a positive modulator of neurotrophin-induced neuronal survival.
Collapse
MESH Headings
- Adaptor Proteins, Vesicular Transport/deficiency
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/metabolism
- Adaptor Proteins, Vesicular Transport/physiology
- Animals
- Axonal Transport/genetics
- Axonal Transport/physiology
- Cell Culture Techniques
- Cerebral Cortex/metabolism
- Embryo, Mammalian/pathology
- Ganglia, Spinal/cytology
- Ganglia, Spinal/metabolism
- HEK293 Cells
- Hippocampus/metabolism
- Humans
- Mice
- Mice, Knockout
- Nerve Growth Factor/pharmacology
- Nerve Growth Factors/physiology
- Neurites/drug effects
- Neurites/physiology
- Receptor Cross-Talk/physiology
- Receptor, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Nerve Growth Factor/physiology
- Signal Transduction/physiology
- Superior Cervical Ganglion/metabolism
- Superior Cervical Ganglion/pathology
Collapse
Affiliation(s)
- Christian B. Vaegter
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Pernille Jansen
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Anja W. Fjorback
- MIND Center, Stereology and Electron Microscopy Laboratory, Aarhus University, 8000 Aarhus C, Denmark
| | - Simon Glerup
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Sune Skeldal
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Mette Richner
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Bettina Erdmann
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Jens R. Nyengaard
- MIND Center, Stereology and Electron Microscopy Laboratory, Aarhus University, 8000 Aarhus C, Denmark
| | - Lino Tessarollo
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, USA
| | - Gary R. Lewin
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | | | - Moses V. Chao
- Kimmel Center at Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York 10016, USA
| | - Anders Nykjaer
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
- NeuronIcon, Gustav Wieds vej 10, DK-8000 Aarhus C, Denmark
| |
Collapse
|
162
|
Liu Y, Abdel Samad O, Zhang L, Duan B, Tong Q, Lopes C, Ji RR, Lowell BB, Ma Q. VGLUT2-dependent glutamate release from nociceptors is required to sense pain and suppress itch. Neuron 2010; 68:543-56. [PMID: 21040853 DOI: 10.1016/j.neuron.2010.09.008] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2010] [Indexed: 12/12/2022]
Abstract
Itch can be suppressed by painful stimuli, but the underlying neural basis is unknown. We generated conditional null mice in which vesicular glutamate transporter type 2 (VGLUT2)-dependent synaptic glutamate release from mainly Nav1.8-expressing nociceptors was abolished. These mice showed deficits in pain behaviors, including mechanical pain, heat pain, capsaicin-evoked pain, inflammatory pain, and neuropathic pain. The pain deficits were accompanied by greatly enhanced itching, as suggested by (1) sensitization of both histamine-dependent and histamine-independent itch pathways and (2) development of spontaneous scratching and skin lesions. Strikingly, intradermal capsaicin injection promotes itch responses in these mutant mice, as opposed to pain responses in control littermates. Consequently, coinjection of capsaicin was no longer able to mask itch evoked by pruritogenic compounds. Our studies suggest that synaptic glutamate release from a group of peripheral nociceptors is required to sense pain and suppress itch. Elimination of VGLUT2 in these nociceptors creates a mouse model of chronic neurogenic itch.
Collapse
Affiliation(s)
- Yang Liu
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, 1 Jimmy Fund Way, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Mishra SK, Tisel SM, Orestes P, Bhangoo SK, Hoon MA. TRPV1-lineage neurons are required for thermal sensation. EMBO J 2010; 30:582-93. [PMID: 21139565 DOI: 10.1038/emboj.2010.325] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 11/15/2010] [Indexed: 01/11/2023] Open
Abstract
The ion-channel TRPV1 is believed to be a major sensor of noxious heat, but surprisingly animals lacking TRPV1 still display marked responses to elevated temperature. In this study, we explored the role of TRPV1-expressing neurons in somatosensation by generating mice wherein this lineage of cells was selectively labelled or ablated. Our data show that TRPV1 is an embryonic marker of many nociceptors including all TRPV1- and TRPM8-neurons as well as many Mrg-expressing neurons. Mutant mice lacking these cells are completely insensitive to hot or cold but in marked contrast retain normal touch and mechanical pain sensation. These animals also exhibit defective body temperature control and lose both itch and pain reactions to potent chemical mediators. Together with previous cell ablation studies, our results define and delimit the roles of TRPV1- and TRPM8-neurons in thermosensation, thermoregulation and nociception, thus significantly extending the concept of labelled lines in somatosensory coding.
Collapse
Affiliation(s)
- Santosh K Mishra
- Molecular Genetics Unit, Laboratory of Sensory Biology, NIDCR, NIH, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
164
|
Zagami CJ, Stifani S. Molecular characterization of the mouse superior lateral parabrachial nucleus through expression of the transcription factor Runx1. PLoS One 2010; 5:e13944. [PMID: 21085653 PMCID: PMC2978708 DOI: 10.1371/journal.pone.0013944] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 10/18/2010] [Indexed: 01/27/2023] Open
Abstract
Background The ability to precisely identify separate neuronal populations is essential to the understanding of the development and function of different brain structures. This necessity is particularly evident in regions such as the brainstem, where the anatomy is quite complex and little is known about the identity, origin, and function of a number of distinct nuclei due to the lack of specific cellular markers. In this regard, the gene encoding the transcription factor Runx1 has emerged as a specific marker of restricted neuronal populations in the murine central and peripheral nervous systems. The aim of this study was to precisely characterize the expression of Runx1 in the developing and postnatal mouse brainstem. Methods and Principal Findings Anatomical and immunohistochemical studies were used to characterize mouse Runx1 expression in the brainstem. It is shown here that Runx1 is expressed in a restricted population of neurons located in the dorsolateral rostral hindbrain. These neurons define a structure that is ventromedial to the dorsal nucleus of the lateral lemniscus, dorsocaudal to the medial paralemniscal nucleus and rostral to the cerebellum. Runx1 expression in these cells is first observed at approximately gestational day 12.5, persists into the adult brain, and is lost in knockout mice lacking the transcription factor Atoh1, an important regulator of the development of neuronal lineages of the rhombic lip. Runx1-expressing neurons in the rostral hindbrain produce cholecystokinin and also co-express members of the Groucho/Transducin-like Enhancer of split protein family. Conclusion Based on the anatomical and molecular characteristics of the Runx1-expressing cells in the rostral hindbrain, we propose that Runx1 expression in this region of the mouse brain defines the superior lateral parabrachial nucleus.
Collapse
Affiliation(s)
- Chrissandra J. Zagami
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Stefano Stifani
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
165
|
Gascon E, Moqrich A. Heterogeneity in primary nociceptive neurons: From molecules to pathology. Arch Pharm Res 2010; 33:1489-507. [DOI: 10.1007/s12272-010-1003-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 08/19/2010] [Accepted: 08/20/2010] [Indexed: 01/17/2023]
|
166
|
Hepatocyte growth factor-Met signaling is required for Runx1 extinction and peptidergic differentiation in primary nociceptive neurons. J Neurosci 2010; 30:12414-23. [PMID: 20844136 DOI: 10.1523/jneurosci.3135-10.2010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nociceptors in peripheral ganglia display a remarkable functional heterogeneity. They can be divided into the following two major classes: peptidergic and nonpeptidergic neurons. Although RUNX1 has been shown to play a pivotal role in the specification of nonpeptidergic neurons, the mechanisms driving peptidergic differentiation remain elusive. Here, we show that hepatocyte growth factor (HGF)-Met signaling acts synergistically with nerve growth factor-tyrosine kinase receptor A to promote peptidergic identity in a subset of prospective nociceptors. We provide in vivo evidence that a population of peptidergic neurons, derived from the RUNX1 lineage, require Met activity for the proper extinction of Runx1 and optimal activation of CGRP (calcitonin gene-related peptide). Moreover, we show that RUNX1 in turn represses Met expression in nonpeptidergic neurons, revealing a bidirectional cross talk between Met and RUNX1. Together, our novel findings support a model in which peptidergic versus nonpeptidergic specification depends on a balance between HGF-Met signaling and Runx1 extinction/maintenance.
Collapse
|
167
|
Nassenstein C, Taylor-Clark TE, Myers AC, Ru F, Nandigama R, Bettner W, Undem BJ. Phenotypic distinctions between neural crest and placodal derived vagal C-fibres in mouse lungs. J Physiol 2010; 588:4769-83. [PMID: 20937710 DOI: 10.1113/jphysiol.2010.195339] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Two major types of nociceptors have been described in dorsal root ganglia (DRGs). In comparison, little is known about the vagal nociceptor subtypes. The vagus nerves provide much of the capsaicin-sensitive nociceptive innervation to visceral tissues, and are likely to contribute to the overall pathophysiology of visceral inflammatory diseases. The cell bodies of these afferent nerves are located in the vagal sensory ganglia referred to as nodose and jugular ganglia. Neurons of the nodose ganglion are derived from the epibranchial placodes, whereas jugular ganglion neurons are derived from the neural crest. In the adult mouse, however, there is often only a single ganglionic structure situated alone in the vagus nerve. By employing Wnt1Cre/R26R mice, which express β-galactosidase only in neural crest derived neurons, we found that this single vagal sensory ganglion is a fused ganglion consisting of both neural crest neurons in the rostral portion and non-neural crest (nodose) neurons in the more central and caudal portions of the structure. Based on their activation and gene expression profiles, we identified two major vagal capsaicin-sensitive nociceptor phenotypes, which innervated a defined target, namely the lung in adult mice. One subtype is non-peptidergic, placodal in origin, expresses P2X2 and P2X3 receptors, responds to α,β-methylene ATP, and expresses TRKB, GFRα1 and RET. The other phenotype is derived from the cranial neural crest and does not express P2X2 receptors and fails to respond to α,β-methylene ATP. This population can be further subdivided into two phenotypes, a peptidergic TRKA(+) and GFRα3(+) subpopulation, and a non-peptidergic TRKB(+) and GFRα1(+) subpopulation. Consistent with their similar embryonic origin, the TRPV1 expressing neurons in the rostral dorsal root ganglia were more similar to jugular than nodose vagal neurons. The data support the hypothesis that vagal nociceptors innervating visceral tissues comprise at least two major subtypes. Due to distinctions in their gene expression profile, each type will respond to noxious or inflammatory conditions in their own unique manner.
Collapse
|
168
|
Liu Y, Ma Q. Generation of somatic sensory neuron diversity and implications on sensory coding. Curr Opin Neurobiol 2010; 21:52-60. [PMID: 20888752 DOI: 10.1016/j.conb.2010.09.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 08/27/2010] [Accepted: 09/03/2010] [Indexed: 11/30/2022]
Abstract
Neurons in the dorsal root ganglia (DRG) are composed of a variety of sensory modalities, three of which are pain-sensing nociceptors, temperature-sensing thermoceptors, and itch-sensing pruriceptors. All these neurons are emerged from a common pool of embryonic DRG neurons that are marked by the expression of the neurotrophin receptor TrkA. Here we discuss how intrinsic transcription factors interface with target-derived signals to specify these functionally distinct sensory neurons. We will also discuss how this control mechanism provides a developmental perspective for the coding of somatic sensations.
Collapse
Affiliation(s)
- Yang Liu
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, 1 Jimmy Fund Way, Boston, MA 02115, USA
| | | |
Collapse
|
169
|
Patil MJ, Jeske NA, Akopian AN. Transient receptor potential V1 regulates activation and modulation of transient receptor potential A1 by Ca2+. Neuroscience 2010; 171:1109-19. [PMID: 20884333 DOI: 10.1016/j.neuroscience.2010.09.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 08/06/2010] [Accepted: 09/20/2010] [Indexed: 11/29/2022]
Abstract
The transient receptor potential A1 (TRPA1) channel contributes to nociceptive signaling in certain pain models. It has been suggested that Ca(2+), which activates and modulates TRPA1, could play a critical regulatory role in this process. Since TRPA1 and transient receptor potential V1 (TRPV1) channels are co-expressed and interact in neurons, we investigated whether activation and modulation of TRPA1 by Ca(2+) is regulated by TRPV1. Cell-attached recordings showed that TRPA1 is activated by extracellular Ca(2+) ([Ca(2+)](e)) in concentration-response fashion. This activation, especially by 2 mM [Ca(2+)](e) was substantially suppressed by co-expression with TRPV1. Inside-out recordings demonstrated that intracellular Ca(2+) ([Ca(2+)](i))-triggered activation of TRPA1 was attenuated by the presence of TRPV1 only at 2 mM [Ca(2+)](e), but not in Ca(2+)-free conditions. Further, depletion of internal Ca(2+) stores by thapsigargin generated TRPA1-mediated currents, which is affected by TRPV1 in both Chinese hamster ovary cells and sensory neurons. Since mustard oil current (I(MO)) is modulated by [Ca(2+)](e), we next examined whether alterations in the Ca(2+)-permeability of TRPV1 by mutating Y671 effect I(MO) properties. First it was demonstrated that the mutations in TRPV1 did not affect association of the TRPA1 and TRPV1 channels. However, these TRPV1 mutations, particularly Y671K, altered the following characteristics of TRPA1: magnitude of I(MO) in presence and absence of [Ca(2+)](e); the influence of [Ca(2+)](e) on the voltage-dependency of I(MO), and open probability of single-channel I(MO). In summary, activation of TRPA1 by [Ca(2+)](e) and [Ca(2+)](i) is controlled by the TRPV1 channel, and characteristics of I(MO) depend on Ca(2+) permeability of the TRPV1 channel.
Collapse
Affiliation(s)
- M J Patil
- Department of Endodontics, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | | | | |
Collapse
|
170
|
da Silva S, Wang F. Retrograde neural circuit specification by target-derived neurotrophins and growth factors. Curr Opin Neurobiol 2010; 21:61-7. [PMID: 20810276 DOI: 10.1016/j.conb.2010.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 07/15/2010] [Accepted: 07/21/2010] [Indexed: 01/13/2023]
Abstract
Neural circuit assembly during development involves a series of highly regulated steps. While genetically pre-determined programs play key roles in the early steps including neurogenesis, migration, and initial growth and guidance of axons; increasing evidence indicates that as the axons reach their targets, the late steps of neuronal differentiation and connectivity formation may be influenced or even specified by target-derived signals. Here we attempt to provide a brief synthesized review on the roles of retrograde neurotrophin and growth factor signaling in regulating the final stages of neural circuit specificity such as axonal projection, dendritic patterning, neurotransmitter phenotype acquisition, and synapse formation.
Collapse
Affiliation(s)
- Susana da Silva
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC 27710, USA
| | | |
Collapse
|
171
|
stac1 and stac2 genes define discrete and distinct subsets of dorsal root ganglia neurons. Gene Expr Patterns 2010; 10:368-75. [PMID: 20736085 DOI: 10.1016/j.gep.2010.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 08/16/2010] [Accepted: 08/19/2010] [Indexed: 11/20/2022]
Abstract
Deciphering the precise in vivo function of a particular neuronal subpopulation is one of the most challenging issues in neurobiology. Dorsal root ganglia (DRG) neurons represent a powerful model system to address this fundamental question. These neurons display many morphological, anatomical and few molecular characteristics. With the aim of expanding the molecular description of the primary sensory neurons, we used Affimetrix microarrays to compare global gene expression profiles of DRG of wild type and trkA(trkC/trkC) knock-in mice at birth and identified several hundred potential markers of nociceptive neurons and few markers of proprioceptive neurons. Here, we describe the identification of two members of a family of putative adapter proteins STAC1 and STAC2. We found STAC1 and STAC2 being expressed in a mutually exclusive fashion in adult DRG neurons. STAC1 mainly marks peptidergic nociceptive neurons while STAC2 is expressed in a subset of nonpeptidergic nociceptors, in all trkB+ neurons and in a subpopulation of proprioceptive neurons. Our expression data demonstrate that STAC proteins identify four categories of primary sensory neurons; one class of peptidergic neurons, a subset of nonpeptidergic neurons, all TrkB+neurons and a subset of proprioceptive neurons. Genetic marking of STACs-expressing sensory neurons will lend significant advance into our understanding of DRG neuronal functional diversity.
Collapse
|
172
|
Abdel Samad O, Liu Y, Yang FC, Kramer I, Arber S, Ma Q. Characterization of two Runx1-dependent nociceptor differentiation programs necessary for inflammatory versus neuropathic pain. Mol Pain 2010; 6:45. [PMID: 20673362 PMCID: PMC2919460 DOI: 10.1186/1744-8069-6-45] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/30/2010] [Indexed: 12/02/2022] Open
Abstract
Background The cellular and molecular programs that control specific types of pain are poorly understood. We reported previously that the runt domain transcription factor Runx1 is initially expressed in most nociceptors and controls sensory neuron phenotypes necessary for inflammatory and neuropathic pain. Results Here we show that expression of Runx1-dependent ion channels and receptors is distributed into two nociceptor populations that are distinguished by persistent or transient Runx1 expression. Conditional mutation of Runx1 at perinatal stages leads to preferential impairment of Runx1-persistent nociceptors and a selective defect in inflammatory pain. Conversely, constitutive Runx1 expression in Runx1-transient nociceptors leads to an impairment of Runx1-transient nociceptors and a selective deficit in neuropathic pain. Notably, the subdivision of Runx1-persistent and Runx1-transient nociceptors does not follow the classical nociceptor subdivision into IB4+ nonpeptidergic and IB4- peptidergic populations. Conclusion Altogether, we have uncovered two distinct Runx1-dependent nociceptor differentiation programs that are permissive for inflammatory versus neuropathic pain. These studies lend support to a transcription factor-based distinction of neuronal classes necessary for inflammatory versus neuropathic pain.
Collapse
Affiliation(s)
- Omar Abdel Samad
- Department of Neurobiology, Dana-Farber Cancer Institute, Harvard Medical School, 1 Jimmy Fund Way, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
173
|
The Mrg Family and Pain*. PROG BIOCHEM BIOPHYS 2010. [DOI: 10.3724/sp.j.1206.2009.00739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
174
|
Takashima Y, Ma L, McKemy DD. The development of peripheral cold neural circuits based on TRPM8 expression. Neuroscience 2010; 169:828-42. [PMID: 20580783 DOI: 10.1016/j.neuroscience.2010.05.039] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 04/27/2010] [Accepted: 05/16/2010] [Indexed: 01/27/2023]
Abstract
Afferent nerve fibers of the somatosensory system are a molecularly diverse cell population that detects a varied range of environmental stimuli, converting these external cues ultimately into a sensory percept. Afferents mediating detection of thermal stimuli express a repertoire of temperature sensitive ion channels of the TRP family which endow these nerves with the ability to respond to the breadth of temperatures in the environment. The cold and menthol receptor TRPM8 is responsible for detection of cold and, unlike other thermosensors, detects both innocuous and noxious temperatures. How this single molecule can perform such diverse functions is currently unknown, but expression analyses in adult tissues shows that TRPM8 neurons are a molecularly diverse population and it is likely that this diversity underlies differential functionality. To determine how this phenotype is established, we examined the developmental time course of TRPM8 expression using a mouse transgenic line in which GFP expression is driven by the TRPM8 transcriptional promoter (Trpm8(GFP)). We find that Trpm8(GFP) expression begins prior to embryonic day 15.5 (E15.5) after which expression reaches levels observed in adult neurons. By E18.5, central axons of Trpm8(GFP) neurons reach the spinal cord dorsal horn, but anatomical localization and in vivo measurements of neural activity suggest that fully functional cold circuits are not established until after the first postnatal week. Additionally, Trpm8(GFP) neurons undergo a transition in neurochemical phenotype, ultimately reaching adult expression of markers such TRPV1, CGRP, peripherin, and NF200 by postnatal day 14. Thus, based on immunochemical, anatomical and functional criteria, active cold neural circuits are fully established by the second week postnatal, thereby suggesting that important extrinsic or intrinsic mechanisms are active prior to this developmental stage.
Collapse
Affiliation(s)
- Y Takashima
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | | | | |
Collapse
|
175
|
Huang T, Liu Y, Huang M, Zhao X, Cheng L. Wnt1-cre-mediated Conditional Loss of Dicer Results in Malformation of the Midbrain and Cerebellum and Failure of Neural Crest and Dopaminergic Differentiation in Mice. J Mol Cell Biol 2010; 2:152-63. [DOI: 10.1093/jmcb/mjq008] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
176
|
Deletion of PIK3C3/Vps34 in sensory neurons causes rapid neurodegeneration by disrupting the endosomal but not the autophagic pathway. Proc Natl Acad Sci U S A 2010; 107:9424-9. [PMID: 20439739 DOI: 10.1073/pnas.0914725107] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The lipid kinase PIK3C3 (also called Vps34) regulates both the endosomal and autophagic pathways. However, the effect of inactivating PIK3C3 on neuronal endosomal versus autophagic processes in vivo has not been studied. We generated mice in which Pik3c3 was conditionally deleted in differentiated sensory neurons. Within a few days after Pik3c3 deletion, mutant large-diameter myelinated neurons accumulated numerous enlarged vacuoles and ubiquitin-positive aggregates and underwent rapid degeneration. By contrast, Pik3c3-deficient small-diameter unmyelinated neurons accumulated excessive numbers of lysosome-like organelles and degenerated more slowly. These differential degenerative phenotypes are unlikely caused by a disruption in the autophagy pathway, because inhibiting autophagy alone by conditional deletion of Atg7 results in a completely distinct phenotype in all sensory neurons (i.e., formation of very large intracellular inclusion bodies and slow degeneration over a period of several months). More surprisingly, a noncanonical PIK3C3-independent LC3-positive autophagosome formation pathway was activated in Pik3c3-deficient small-diameter neurons. Analyses of Pik3c3/Atg7 double mutant neurons revealed that this unconventional initiation pathway still depends on ATG7. Our studies represent in vivo characterization of PIK3C3 functions in mammals and provide insights into the complexity of neuronal endo-lysosomal and autophagic pathways.
Collapse
|
177
|
TRPV2 enhances axon outgrowth through its activation by membrane stretch in developing sensory and motor neurons. J Neurosci 2010; 30:4601-12. [PMID: 20357111 DOI: 10.1523/jneurosci.5830-09.2010] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Thermosensitive TRP (thermo TRP) channels are well recognized for their contributions to sensory transduction, responding to a wide variety of stimuli including temperature, nociceptive stimuli, touch, and osmolarity. However, the precise roles for the thermo TRP channels during development have not been determined. To explore the functional importance of thermo TRP channels during neural development, the temporal expression was determined in embryonic mice. Interestingly, TRPV2 expression was detected in spinal motor neurons in addition to the dorsal root ganglia from embryonic day 10.5 and was localized in axon shafts and growth cones, suggesting that the channel is important for axon outgrowth regulation. We revealed that endogenous TRPV2 was activated in a membrane stretch-dependent manner in developing neurons by knocking down the TRPV2 function with dominant-negative TRPV2 and TRPV2-specific shRNA and significantly promoted axon outgrowth. Thus, for the first time we revealed that TRPV2 is an important regulator for axon outgrowth through its activation by membrane stretch during development.
Collapse
|
178
|
Gregus AM, Inra CN, Giordano TP, Costa ACS, Rajadhyaksha AM, Inturrisi CE. Spinal mediators that may contribute selectively to antinociceptive tolerance but not other effects of morphine as revealed by deletion of GluR5. Neuroscience 2010; 169:475-87. [PMID: 20359526 DOI: 10.1016/j.neuroscience.2010.03.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 03/23/2010] [Accepted: 03/23/2010] [Indexed: 12/22/2022]
Abstract
Several groups maintain that morphine tolerance and dependence correlate with increased activity of protein kinases ERK1/2 and P38 MAPK and PKC as well as elevated levels of the neuropeptides dynorphin (DYN), substance P (sP), and calcitonin gene-related peptide (CGRP) in spinal cord dorsal horn (SCDH). They demonstrate that tolerance and dependence can be prevented, and sometimes reversed, by constitutive genetic deletion or pharmacological inhibition of these factors. Recently, we showed that mice with a constitutive deletion of the GluR5 subunit of kainate receptors (GluR5 KO) are not different from wild type (WT) littermates with respect to baseline nociceptive thresholds as well as acute morphine antinociception, morphine physical dependence and conditioned place preference. However, unlike WT, GluR5 KO mice do not develop antinociceptive tolerance following systemic morphine administration. In this report, we examined levels of these mediators in SCDH of WT and GluR5 KO mice following subcutaneous implantation of placebo or morphine pellets. Surprisingly, spinal DYN and CGRP, along with phosphorylated ERK2 (pERK2), P38 (pP38) and PKCgamma (pPKCgamma) are elevated by deletion of GluR5. Additionally, chronic systemic morphine administration increased spinal pERK2, pP38 and pPKCgamma levels in both tolerant WT and non-tolerant GluR5 KO mice. In contrast, while morphine increased spinal DYN and CGRP in WT mice, DYN remained unchanged and CGRP was reduced in GluR5 KO mice. These observations suggest that spinal ERK2, P38 and PKCgamma are likely involved in multiple adaptive responses following systemic morphine administration, whereas DYN and CGRP may contribute selectively to the development of antinociceptive tolerance.
Collapse
Affiliation(s)
- A M Gregus
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | |
Collapse
|
179
|
George L, Kasemeier-Kulesa J, Nelson BR, Koyano-Nakagawa N, Lefcort F. Patterned assembly and neurogenesis in the chick dorsal root ganglion. J Comp Neurol 2010; 518:405-22. [PMID: 20017208 DOI: 10.1002/cne.22248] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The birth of small-diameter TrkA+ neurons that mediate pain and thermoreception begins approximately 24 hours after the cessation of neural crest cell migration from progenitors residing in the nascent dorsal root ganglion. Although multiple geographically distinct progenitor pools have been proposed, this study is the first to comprehensively characterize the derivation of small-diameter neurons. In the developing chick embryo we identify novel patterns in neural crest cell migration and colonization that sculpt the incipient ganglion into a postmitotic neuronal core encapsulated by a layer of proliferative progenitor cells. Furthermore, we show that this outer progenitor layer is composed of three spatially, temporally, and molecularly distinct progenitor zones, two of which give rise to distinct populations of TrkA+ neurons.
Collapse
Affiliation(s)
- Lynn George
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana 59717, USA.
| | | | | | | | | |
Collapse
|
180
|
Runx1 directly promotes proliferation of hair follicle stem cells and epithelial tumor formation in mouse skin. Mol Cell Biol 2010; 30:2518-36. [PMID: 20308320 DOI: 10.1128/mcb.01308-09] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Runx1/AML1 is a transcription factor implicated in tissue stem cell regulation and belongs to the small Runx family of cancer genes. In the hair follicle (HF), Runx1 epithelial deletion in morphogenesis impairs normal adult hair homeostasis (cycle) and blocks adult hair follicle stem cells (HFSCs) in quiescence. Here, we show that these effects are overcome later in adulthood. By deleting Runx1 after the end of morphogenesis, we demonstrate its direct role in promoting anagen onset and HFSC proliferation. Runx1 deletion resulted in cyclin-dependent kinase inhibitor Cdkn1a (p21) upregulation. Interfering with Runx1 function in cultured HFSCs impaired their proliferation and normal G(0)/G1 and G(1)/S cell cycle progression. The proliferation defect could be rescued by Runx1 readdition or by p21 deletion. Chemically induced skin tumorigenesis in mice turned on broad Runx1 expression in regions of the skin epithelium, papillomas, and squamous cell carcinomas. In addition, it revealed reduced rates of tumor formation in the absence of Runx1 that were accompanied by decreased epithelial levels of phospho-Stat3. Runx1 protein expression was similar in normal human and mouse hair cycles. We propose that Runx1 may act as a skin oncogene by directly promoting proliferation of the epithelial cells.
Collapse
|
181
|
Lanier J, Dykes IM, Nissen S, Eng SR, Turner EE. Brn3a regulates the transition from neurogenesis to terminal differentiation and represses non-neural gene expression in the trigeminal ganglion. Dev Dyn 2010; 238:3065-79. [PMID: 19877281 DOI: 10.1002/dvdy.22145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The POU-domain transcription factor Brn3a is expressed in developing sensory neurons at all levels of the neural axis, including the trigeminal ganglion, hindbrain sensory ganglia, and dorsal root ganglia. Changes in global gene expression in the trigeminal ganglion from E11.5 to E13.5 reflect the repression of early neurogenic genes, exit from the cell cycle, and initiation of the expression of definitive markers of sensory function. A majority of these developmental changes are perturbed in the trigeminal ganglia of Brn3a knockout mice. At E13.5, Brn3a(-/-) trigeminal neurons fail to repress a battery of developmental regulators that are highly expressed at E11.5 and are normally down-regulated as development progresses, and also fail to appropriately activate a set of definitive sensory genes. Remarkably, developing Brn3a(-/-) trigeminal neurons also ectopically express multiple regulatory genes associated with cardiac and/or cranial mesoderm development, although definitive myogenic programs are not activated. The majority of these genes are not ectopically expressed in the dorsal root ganglia of Brn3a null mice, perhaps due to redundant mechanisms of repression at spinal levels. These results underscore the importance of gene repression in regulating neuronal development, and the need for unbiased screens in the determination of developmental gene regulatory programs.
Collapse
Affiliation(s)
- Jason Lanier
- Department of Psychiatry and University of California, San Diego, California 92093-0603, USA
| | | | | | | | | |
Collapse
|
182
|
Philipot O, Joliot V, Ait-Mohamed O, Pellentz C, Robin P, Fritsch L, Ait-Si-Ali S. The core binding factor CBF negatively regulates skeletal muscle terminal differentiation. PLoS One 2010; 5:e9425. [PMID: 20195544 PMCID: PMC2828485 DOI: 10.1371/journal.pone.0009425] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 02/03/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Core Binding Factor or CBF is a transcription factor composed of two subunits, Runx1/AML-1 and CBF beta or CBFbeta. CBF was originally described as a regulator of hematopoiesis. METHODOLOGY/PRINCIPAL FINDINGS Here we show that CBF is involved in the control of skeletal muscle terminal differentiation. Indeed, downregulation of either Runx1 or CBFbeta protein level accelerates cell cycle exit and muscle terminal differentiation. Conversely, overexpression of CBFbeta in myoblasts slows terminal differentiation. CBF interacts directly with the master myogenic transcription factor MyoD, preferentially in proliferating myoblasts, via Runx1 subunit. In addition, we show a preferential recruitment of Runx1 protein to MyoD target genes in proliferating myoblasts. The MyoD/CBF complex contains several chromatin modifying enzymes that inhibits MyoD activity, such as HDACs, Suv39h1 and HP1beta. When overexpressed, CBFbeta induced an inhibition of activating histone modification marks concomitant with an increase in repressive modifications at MyoD target promoters. CONCLUSIONS/SIGNIFICANCE Taken together, our data show a new role for Runx1/CBFbeta in the control of the proliferation/differentiation in skeletal myoblasts.
Collapse
Affiliation(s)
- Ophélie Philipot
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
| | - Véronique Joliot
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
| | - Ouardia Ait-Mohamed
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
| | - Céline Pellentz
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
| | - Philippe Robin
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
| | - Lauriane Fritsch
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
| | - Slimane Ait-Si-Ali
- Institut André Lwoff, FRE2944, CNRS and Université Paris-Sud, Villejuif, France
- * E-mail:
| |
Collapse
|
183
|
Bourane S, Garces A, Venteo S, Pattyn A, Hubert T, Fichard A, Puech S, Boukhaddaoui H, Baudet C, Takahashi S, Valmier J, Carroll P. Low-threshold mechanoreceptor subtypes selectively express MafA and are specified by Ret signaling. Neuron 2010; 64:857-70. [PMID: 20064392 DOI: 10.1016/j.neuron.2009.12.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2009] [Indexed: 02/01/2023]
Abstract
Low-threshold mechanoreceptor neurons (LTMs) of the dorsal root ganglia (DRG) are essential for touch sensation. They form highly specialized terminations in the skin and display stereotyped projections in the spinal cord. Functionally defined LTMs depend on neurotrophin signaling for their postnatal survival and functioning, but how these neurons arise during development is unknown. Here, we show that specific types of LTMs can be identified shortly after DRG genesis by unique expression of the MafA transcription factor, the Ret receptor and coreceptor GFRalpha2, and find that their specification is Ngn2 dependent. In mice lacking Ret, these LTMs display early differentiation defects, as revealed by reduced MafA expression, and at later stages their central and peripheral projections are compromised. Moreover, in MafA mutants, a discrete subset of LTMs display altered expression of neurotrophic factor receptors. Our results provide evidence that genetic interactions involving Ret and MafA progressively promote the differentiation and diversification of LTMs.
Collapse
|
184
|
Linley JE, Rose K, Ooi L, Gamper N. Understanding inflammatory pain: ion channels contributing to acute and chronic nociception. Pflugers Arch 2010; 459:657-69. [DOI: 10.1007/s00424-010-0784-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 01/04/2010] [Accepted: 01/06/2010] [Indexed: 02/06/2023]
|
185
|
Development of multilineage adult hematopoiesis in the zebrafish with a runx1 truncation mutation. Blood 2010; 115:2806-9. [PMID: 20154212 DOI: 10.1182/blood-2009-08-236729] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Runx1 is required for the emergence of hematopoietic stem cells (HSCs) from hemogenic endothelium during embryogenesis. However, its role in the generation and maintenance of HSCs during adult hematopoiesis remains uncertain. Here, we present analysis of a zebrafish mutant line carrying a truncation mutation, W84X, in runx1. The runx1(W84X/W84X) embryos showed blockage in the initiation of definitive hematopoiesis, but some embryos were able to recover from a larval "bloodless" phase and develop to fertile adults with multilineage hematopoiesis. Using cd41-green fluorescent protein transgenic zebrafish and lineage tracing, we demonstrated that the runx1(W84X/W84X) embryos developed cd41(+) HSCs in the aorta-gonad-mesonephros region, which later migrated to the kidney, the site of adult hematopoiesis. Overall, our data suggest that in zebrafish adult HSCs can be formed without an intact runx1.
Collapse
|
186
|
Kanaykina N, Abelson K, King D, Liakhovitskaia A, Schreiner S, Wegner M, Kozlova EN. In vitro and in vivo effects on neural crest stem cell differentiation by conditional activation of Runx1 short isoform and its effect on neuropathic pain behavior. Ups J Med Sci 2010; 115:56-64. [PMID: 20187849 PMCID: PMC2853355 DOI: 10.3109/03009730903572065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Runx1, a Runt domain transcription factor, controls the differentiation of nociceptors that express the neurotrophin receptor Ret, regulates the expression of many ion channels and receptors, and controls the lamina-specific innervation pattern of nociceptive afferents in the spinal cord. Moreover, mice lacking Runx1 exhibit specific defects in thermal and neuropathic pain. We investigated whether conditional activation of Runx1 short isoform (Runx1a), which lacks a transcription activation domain, influences differentiation of neural crest stem cells (NCSCs) in vitro and in vivo during development and whether postnatal Runx1a activation affects the sensitivity to neuropathic pain. METHODS We activated ectopic expression of Runx1a in cultured NCSCs using the Tet-ON gene regulatory system during the formation of neurospheres and analyzed the proportion of neurons and glial cells originating from NCSCs. In in vivo experiments we applied doxycycline (DOX) to pregnant mice (days 8-11), i.e. when NCSCs actively migrate, and examined the phenotype of offsprings. We also examined whether DOX-induced activation of Runx1a in adult mice affects their sensitivity to mechanical stimulation following a constriction injury of the sciatic nerve. RESULTS Ectopic Runx1a expression in cultured NCSCs resulted in predominantly glial differentiation. Offsprings in which Runx1a had been activated showed retarded growth and displayed megacolon, pigment defects, and dystrophic dorsal root ganglia. In the neuropathic pain model, the threshold for mechanical sensitivity was markedly increased following activation of Runx1a. CONCLUSION These data suggest that Runx1a has a specific role in NCSC development and that modulation of Runx1a activity may reduce mechanical hypersensitivity associated with neuropathic pain.
Collapse
Affiliation(s)
- Nadezda Kanaykina
- Department of Neuroscience, Neuroanatomy, Uppsala University Biomedical Center, UppsalaSweden
| | | | - Dale King
- Department of Neuroscience, Neuroanatomy, Uppsala University Biomedical Center, UppsalaSweden
| | | | - Silke Schreiner
- Department of Biochemistry, University of Erlangen-Nuremberg, ErlangenGermany
| | - Michael Wegner
- Department of Biochemistry, University of Erlangen-Nuremberg, ErlangenGermany
| | - Elena N. Kozlova
- Department of Neuroscience, Neuroanatomy, Uppsala University Biomedical Center, UppsalaSweden
| |
Collapse
|
187
|
Dykes IM, Lanier J, Eng SR, Turner EE. Brn3a regulates neuronal subtype specification in the trigeminal ganglion by promoting Runx expression during sensory differentiation. Neural Dev 2010; 5:3. [PMID: 20096094 PMCID: PMC2829025 DOI: 10.1186/1749-8104-5-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 01/22/2010] [Indexed: 01/03/2023] Open
Abstract
The transcription factor Brn3a, product of the pou4f1 gene, is expressed in most sensory neurons throughout embryogenesis. Prior work has demonstrated a role for Brn3a in the repression of early neurogenic genes; here we describe a second major role for Brn3a in the specification of sensory subtypes in the trigeminal ganglion (TG). Sensory neurons initially co-express multiple Trk-family neurotrophin receptors, but are later marked by the unique expression of TrkA, TrkB or TrkC. Maturation of these sensory subtypes is known to depend on the expression of Runx transcription factors. Newborn Brn3a knockout mice fail to express TrkC, which is associated in the TG with mechanoreceptors, plus a set of functional genes associated with nociceptor subtypes. In embryonic Brn3a-/- ganglia, the normal expression of Runx3 is never initiated in TrkC+ neurons, and Runx1 expression is greatly attenuated in TrkA+ nociceptors. These changes are accompanied by expanded expression of TrkB in neurons that abnormally express multiple Trks, followed by the loss of TrkC and TrkA expression. In transgenic embryos expressing a Brn3a-VP16 dominant transactivator, Runx3 mRNA expression is increased, suggesting that it is a direct regulatory target of Brn3a. Chromatin immunoprecipitation confirms that Brn3a binds in vivo to a conserved upstream enhancer element within histone H3-acetylated chromatin in the Runx3 locus. Together these data show that Brn3a acts upstream of the Runx factors, which then repress TrkB expression to allow establishment of the non-overlapping Trk receptor profiles and correct terminally differentiated phenotypes.
Collapse
Affiliation(s)
- Iain M Dykes
- Department of Psychiatry, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0603, USA
| | | | | | | |
Collapse
|
188
|
Zagami CJ, Zusso M, Stifani S. Runx transcription factors: lineage-specific regulators of neuronal precursor cell proliferation and post-mitotic neuron subtype development. J Cell Biochem 2009; 107:1063-72. [PMID: 19507194 DOI: 10.1002/jcb.22221] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Runt-related (RUNX) genes encode evolutionarily conserved transcription factors that play essential roles during development and adult tissue homeostasis. RUNX proteins regulate the transition from proliferation to differentiation in a variety of cell lineages. Moreover, they control the diversification of distinct cellular phenotypes in numerous tissues. Alterations of RUNX functions are associated with several cancers and other human pathologies, underscoring the vital roles of these transcription factors in adult organs. Insights into the functions and regulations of mammalian RUNX proteins have been provided mostly by studies of RUNX involvement in mechanisms of hematopoietic and skeletal development and disease. A growing number of recent investigations are revealing new functions for RUNX family members during the development of the mammalian nervous system. This review will discuss recent progress in the study of RUNX protein involvement in mammalian neural development, with emphasis on the differentiation of olfactory, sensory, and motor neuron lineages.
Collapse
|
189
|
|
190
|
Kostyuk EP, Kostyuk PG. Peculiarities of Ion Channels and Modulation of Their Functions in Neurons Belonging to the Nociceptive System. NEUROPHYSIOLOGY+ 2009. [DOI: 10.1007/s11062-009-9090-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
191
|
Glover JC. "The developmental and functional logic of neuronal circuits": commentary on the Kavli Prize in Neuroscience. Neuroscience 2009; 163:977-84. [PMID: 19664740 DOI: 10.1016/j.neuroscience.2009.07.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 07/13/2009] [Accepted: 07/24/2009] [Indexed: 11/27/2022]
Abstract
The first Kavli Prize in Neuroscience recognizes a confluence of career achievements that together provide a fundamental understanding of how brain and spinal cord circuits are assembled during development and function in the adult. The members of the Kavli Neuroscience Prize Committee have decided to reward three scientists (Sten Grillner, Thomas Jessell, and Pasko Rakic) jointly "for discoveries on the developmental and functional logic of neuronal circuits". Pasko Rakic performed groundbreaking studies of the developing cerebral cortex, including the discovery of how radial glia guide the neuronal migration that establishes cortical layers and for the radial unit hypothesis and its implications for cortical connectivity and evolution. Thomas Jessell discovered molecular principles governing the specification and patterning of different neuron types and the development of their synaptic interconnection into sensorimotor circuits. Sten Grillner elucidated principles of network organization in the vertebrate locomotor central pattern generator, along with its command systems and sensory and higher order control. The discoveries of Rakic, Jessell and Grillner provide a framework for how neurons obtain their identities and ultimate locations, establish appropriate connections with each other, and how the resultant neuronal networks operate. Their work has significantly advanced our understanding of brain development and function and created new opportunities for the treatment of neurological disorders. Each has pioneered an important area of neuroscience research and left a legacy of exceptional scientific achievement, insight, communication, mentoring and leadership.
Collapse
Affiliation(s)
- J C Glover
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Blindern, Oslo, Norway
| |
Collapse
|
192
|
Aldskogius H, Berens C, Kanaykina N, Liakhovitskaia A, Medvinsky A, Sandelin M, Schreiner S, Wegner M, Hjerling-Leffler J, Kozlova EN. Regulation of boundary cap neural crest stem cell differentiation after transplantation. Stem Cells 2009; 27:1592-603. [PMID: 19544468 PMCID: PMC2733376 DOI: 10.1002/stem.77] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Success of cell replacement therapies for neurological disorders will depend largely on the optimization of strategies to enhance viability and control the developmental fate of stem cells after transplantation. Once transplanted, stem/progenitor cells display a tendency to maintain an undifferentiated phenotype or differentiate into inappropriate cell types. Gain and loss of function experiments have revealed key transcription factors which drive differentiation of immature stem/progenitor cells toward more mature stages and eventually to full differentiation. An attractive course of action to promote survival and direct the differentiation of transplanted stem cells to a specific cell type would therefore be to force expression of regulatory differentiation molecules in already transplanted stem cells, using inducible gene expression systems which can be controlled from the outside. Here, we explore this hypothesis by employing a tetracycline gene regulating system (Tet-On) to drive the differentiation of boundary cap neural crest stem cells (bNCSCs) toward a sensory neuron fate after transplantation. We induced the expression of the key transcription factor Runx1 in Sox10-expressing bNCSCs. Forced expression of Runx1 strongly increased transplant survival in the enriched neurotrophic environment of the dorsal root ganglion cavity, and was sufficient to guide differentiation of bNCSCs toward a nonpeptidergic nociceptive sensory neuron phenotype both in vitro and in vivo after transplantation. These findings suggest that exogenous activation of transcription factors expression after transplantation in stem/progenitor cell grafts can be a constructive approach to control their survival as well as their differentiation to the desired type of cell and that the Tet-system is a useful tool to achieve this.
Collapse
Affiliation(s)
- Hakan Aldskogius
- Department of Neuroscience, Neuroanatomy, Uppsala University Biomedical Center, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Staaf S, Franck MCM, Marmigère F, Mattsson JP, Ernfors P. Dynamic expression of the TRPM subgroup of ion channels in developing mouse sensory neurons. Gene Expr Patterns 2009; 10:65-74. [PMID: 19850157 DOI: 10.1016/j.gep.2009.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 09/10/2009] [Accepted: 10/12/2009] [Indexed: 12/24/2022]
Abstract
Despite the significance of transient receptor potential (TRP) channels in sensory physiology, little is known of the expression and developmental regulation of the TRPM (melastatin) subgroup in sensory neurons. In order to find out if the eight TRPM subgroup members (TRPM1-TRPM8) have a possible role in the sensory nervous system, we characterized the developmental regulation of their expression in mouse dorsal root ganglion (DRG) from embryonic (E) day 12 to adulthood. Transcripts for all channels except for TRPM1 were detected in lumbar and thoracic DRG and in nodose ganglion (NG) with distinguishable expression patterns from E12 until adult. For most channels, the expression increased from E14 to adult with the exception of TRPM5, which displayed transient high levels during embryonic and early postnatal stages. Cellular localization of TRPM8 mRNA was found only in a limited subset of very small diameter neurons distinct in size from other populations. These neurons did not bind isolectin B4 (IB4) and expressed neither the neuropeptide calcitonin gene-related peptide (CGRP) nor neurofilament (NF)200. This suggests that TRPM8(+) thermoreceptive sensory neurons fall into a separate group of very small sized neurons distinct from peptidergic and IB4(+) subtypes of sensory neurons. Our results, showing the expression and dynamic regulation of TRPM channels during development, indicate that many TRPM subfamily members could participate during nervous system development and in the adult by determining distinct physiological properties of sensory neurons.
Collapse
Affiliation(s)
- Susanne Staaf
- Department of Bioscience, AstraZeneca R&D Mölndal, 431 83 Mölndal, Sweden
| | | | | | | | | |
Collapse
|
194
|
Abstract
P2X and P2Y nucleotide receptors are described on sensory neurons and their peripheral and central terminals in dorsal root, nodose, trigeminal, petrosal, retinal and enteric ganglia. Peripheral terminals are activated by ATP released from local cells by mechanical deformation, hypoxia or various local agents in the carotid body, lung, gut, bladder, inner ear, eye, nasal organ, taste buds, skin, muscle and joints mediating reflex responses and nociception. Purinergic receptors on fibres in the dorsal spinal cord and brain stem are involved in reflex control of visceral and cardiovascular activity, as well as relaying nociceptive impulses to pain centres. Purinergic mechanisms are enhanced in inflammatory conditions and may be involved in migraine, pain, diseases of the special senses, bladder and gut, and the possibility that they are also implicated in arthritis, respiratory disorders and some central nervous system disorders is discussed. Finally, the development and evolution of purinergic sensory mechanisms are considered.
Collapse
|
195
|
Gonzalez-Brito MR, Bixby JL. Protein tyrosine phosphatase receptor type O regulates development and function of the sensory nervous system. Mol Cell Neurosci 2009; 42:458-65. [PMID: 19800005 DOI: 10.1016/j.mcn.2009.09.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 09/04/2009] [Accepted: 09/21/2009] [Indexed: 12/24/2022] Open
Abstract
The roles of protein tyrosine phosphatases (PTPs) in differentiation and axon targeting by dorsal root ganglion (DRG) neurons are essentially unknown. The type III transmembrane PTP, PTPRO, is expressed in DRG neurons, and is implicated in the guidance of motor and retinal axons. We examined the role of PTPRO in DRG development and function using PTPRO(-/-) mice. The number of peptidergic nociceptive neurons in the DRG of PTPRO(-/-) mice was significantly decreased, while the total number of sensory neurons appeared unchanged. In addition, spinal pathfinding by both peptidergic and proprioceptive neurons was abnormal in PTPRO(-/-) mice. Lastly, PTPRO(-/-) mice performed abnormally on tests of thermal pain and sensorimotor coordination, suggesting that both nociception and proprioception were perturbed. Our data indicate that PTPRO is required for peptidergic differentiation and process outgrowth of sensory neurons, as well as mature sensory function, and provide the first evidence that RPTPs regulate DRG development.
Collapse
Affiliation(s)
- Manuel R Gonzalez-Brito
- Department of Pediatrics, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Lois Pope LIFE Center, Room 4-17, 1095 Northwest 14th Terrace, Miami, FL 33136, USA
| | | |
Collapse
|
196
|
Mandai K, Guo T, Hillaire CS, Meabon JS, Kanning KC, Bothwell M, Ginty DD. LIG family receptor tyrosine kinase-associated proteins modulate growth factor signals during neural development. Neuron 2009; 63:614-27. [PMID: 19755105 PMCID: PMC2758028 DOI: 10.1016/j.neuron.2009.07.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 06/04/2009] [Accepted: 07/25/2009] [Indexed: 01/19/2023]
Abstract
Genome-wide screens were performed to identify transmembrane proteins that mediate axonal growth, guidance and target field innervation of somatosensory neurons. One gene, Linx (alias Islr2), encoding a leucine-rich repeat and immunoglobulin (LIG) family protein, is expressed in a subset of developing sensory and motor neurons. Domain and genomic structures of Linx and other LIG family members suggest that they are evolutionarily related to Trk receptor tyrosine kinases (RTKs). Several LIGs, including Linx, are expressed in subsets of somatosensory and motor neurons, and select members interact with TrkA and Ret RTKs. Moreover, axonal projection defects in mice harboring a null mutation in Linx resemble those in mice lacking Ngf, TrkA, and Ret. In addition, Linx modulates NGF-TrkA- and GDNF-GFRalpha1/Ret-mediated axonal extension in cultured sensory and motor neurons, respectively. These findings show that LIGs physically interact with RTKs and modulate their activities to control axonal extension, guidance and branching.
Collapse
Affiliation(s)
- Kenji Mandai
- The Solomon H. Snyder Department of Neuroscience, The Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, PCTB 1015, Baltimore, MD 21205, USA
| | - Ting Guo
- The Solomon H. Snyder Department of Neuroscience, The Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, PCTB 1015, Baltimore, MD 21205, USA
| | - Coryse St. Hillaire
- The Solomon H. Snyder Department of Neuroscience, The Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, PCTB 1015, Baltimore, MD 21205, USA
| | - James S. Meabon
- Department of Physiology and Biophysics, Box 357290, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Kevin C. Kanning
- Department of Physiology and Biophysics, Box 357290, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - Mark Bothwell
- Department of Physiology and Biophysics, Box 357290, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | - David D. Ginty
- The Solomon H. Snyder Department of Neuroscience, The Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, PCTB 1015, Baltimore, MD 21205, USA
| |
Collapse
|
197
|
|
198
|
Loss of the putative catalytic domain of HDAC4 leads to reduced thermal nociception and seizures while allowing normal bone development. PLoS One 2009; 4:e6612. [PMID: 19672313 PMCID: PMC2720538 DOI: 10.1371/journal.pone.0006612] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Accepted: 07/06/2009] [Indexed: 12/02/2022] Open
Abstract
Histone deacetylase 4 (HDAC4) has been associated with muscle & bone development [1]–[6]. N-terminal MEF2 and RUNX2 binding domains of HDAC4 have been shown to mediate these effects in vitro. A complete gene knockout has been reported to result in premature ossification and associated defects resulting in postnatal lethality [6]. We report a viral insertion mutation that deletes the putative deacetylase domain, while preserving the N-terminal portion of the protein. Western blot and immuno-precipitation analysis confirm expression of truncated HDAC4 containing N-terminal amino acids 1-747. These mutant mice are viable, living to at least one year of age with no gross defects in muscle or bone. At 2–4 months of age no behavioral or physiological abnormalities were detected except for an increased latency to respond to a thermal nociceptive stimulus. As the mutant mice aged past 5 months, convulsions appeared, often elicited by handling. Our findings confirm the sufficiency of the N-terminal domain for muscle and bone development, while revealing other roles of HDAC4.
Collapse
|
199
|
Tsunozaki M, Bautista DM. Mammalian somatosensory mechanotransduction. Curr Opin Neurobiol 2009; 19:362-9. [PMID: 19683913 PMCID: PMC4044613 DOI: 10.1016/j.conb.2009.07.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 07/10/2009] [Accepted: 07/13/2009] [Indexed: 11/22/2022]
Abstract
In the mammalian somatosensory system, mechanosensitive neurons mediate the senses of touch and pain. Among sensory modalities, mechanosensation has been the most elusive with regard to the identification of transduction molecules. One factor that has hindered the identification of transduction molecules is the diversity of neurons; physiological studies have revealed many subtypes of neurons, specialized to detect a variety of mechanical stimuli. Do different subtypes use the same transduction molecules that are modified by cellular context? Or, are there multiple mechanotransducers that specialize in sensing different mechanical stimuli? This review highlights recent progress in identifying and characterizing candidate molecular force transducers, as well as the development of new tools to characterize touch transduction at the molecular, cellular, and behavioral levels.
Collapse
Affiliation(s)
- Makoto Tsunozaki
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | | |
Collapse
|
200
|
Macrophage stimulating protein is a neurotrophic factor for a sub-population of adult nociceptive sensory neurons. Mol Cell Neurosci 2009; 41:175-85. [DOI: 10.1016/j.mcn.2009.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 12/31/2022] Open
|