51
|
Abstract
NTRK gene fusions involving either NTRK1, NTRK2 or NTRK3 (encoding the neurotrophin receptors TRKA, TRKB and TRKC, respectively) are oncogenic drivers of various adult and paediatric tumour types. These fusions can be detected in the clinic using a variety of methods, including tumour DNA and RNA sequencing and plasma cell-free DNA profiling. The treatment of patients with NTRK fusion-positive cancers with a first-generation TRK inhibitor, such as larotrectinib or entrectinib, is associated with high response rates (>75%), regardless of tumour histology. First-generation TRK inhibitors are well tolerated by most patients, with toxicity profiles characterized by occasional off-tumour, on-target adverse events (attributable to TRK inhibition in non-malignant tissues). Despite durable disease control in many patients, advanced-stage NTRK fusion-positive cancers eventually become refractory to TRK inhibition; resistance can be mediated by the acquisition of NTRK kinase domain mutations. Fortunately, certain resistance mutations can be overcome by second-generation TRK inhibitors, including LOXO-195 and TPX-0005 that are being explored in clinical trials. In this Review, we discuss the biology of NTRK fusions, strategies to target these drivers in the treatment-naive and acquired-resistance disease settings, and the unique safety profile of TRK inhibitors.
Collapse
|
52
|
Li Z, Rasmussen TS, Rasmussen ML, Li J, Henríquez Olguín C, Kot W, Nielsen DS, Jensen TE. The Gut Microbiome on a Periodized Low-Protein Diet Is Associated With Improved Metabolic Health. Front Microbiol 2019; 10:709. [PMID: 31019501 PMCID: PMC6458274 DOI: 10.3389/fmicb.2019.00709] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/21/2019] [Indexed: 12/15/2022] Open
Abstract
A periodized (14 days on/14 days off) 5% low protein-high carbohydrate (pLPHC) diet protects against weight gain, improves glucose tolerance in mice and interacts with concurrent voluntary activity wheel training on several parameters including weight maintenance and liver FGF21 secretion. The gut microbiome (GM) responds to both diet and exercise and may influence host metabolism. This study compared the cecal GM after a 13.5-week intervention study in mice on a variety of dietary interventions ± concurrent voluntary exercise training in activity wheels. The diets included chronic chow diet, LPHC diet, 40 E% high protein-low carbohydrate (HPLC) diet, an obesigenic chronic high-fat diet (HFD) and the pLPHC diet. Our hypothesis was that the GM changes with pLPHC diet would generally reflect the improved metabolic health of the host and interact with concurrent exercise training. The GM analyses revealed greater abundance phylum Bacteroidetes and the genus Akkermansia on chronic and periodized LPHC and higher abundance of Oscillospira and Oscillibacter on HFD. The differences in diet-induced GM correlated strongly with the differences in a range of host metabolic health-measures. In contrast, no significant effect of concurrent exercise training was observed. In conclusion, pLPHC diet elicits substantial changes in the GM. In contrast, only subtle and non-significant effects of concurrent activity wheel exercise were observed. The pLPHC-associated microbiome may contribute to the healthier host phenotype observed in these mice.
Collapse
Affiliation(s)
- Zhencheng Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Torben Sølbeck Rasmussen
- Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Mette Line Rasmussen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jingwen Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Henríquez Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Witold Kot
- Department of Environmental Sciences, Aarhus University, Roskilde, Denmark
| | - Dennis Sandris Nielsen
- Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Thomas Elbenhardt Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
53
|
Wang Y, Wang J, Wang H, Feng X, Tao Y, Yang J, Cai J. Tet1 Overexpression and Decreased DNA Hydroxymethylation Protect Neurons Against Cell Death After Injury by Increasing Expression of Genes Involved in Cell Survival. World Neurosurg 2019; 126:e713-e722. [PMID: 30849555 DOI: 10.1016/j.wneu.2019.02.133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Spinal cord and neuron injury result in loss of muscle function, sensation, or autonomic function in the body. Tet1 produces 5-hydroxymethylcytosin. The conversion was proposed as the initial step of deoxyribonucleic acid demethylation in mammals. However, effects of Tet1 expression and hydroxymethylation status on neuron injury remain unclear. Therefore the current study was designed to explore effects of Tet1 expression and hydroxymethylation status on cell survival and gene expression after neuron injury. METHODS Mouse models of spinal cord injury and cell model of neuron injury were created. Animals were sacrificed, and injured spinal cord tissue was harvested. Neuron-like cells were cultured after scratch injury. Hydroxymethylated deoxyribonucleic acid concentration was detected, and Tet1 expression was examined by qPCR. Neuron-like cells were divided into 3 groups: control, injury, and azacytidine + injury (before injury, cells were pretreated with azacytidine) groups. Culture supernatant was collected, and lactate dehydrogenase concentration was detected. Meanwhile, injured neuron-like cells were divided into 3 groups: negative control, Tet1 overexpression, and Tet1 interference. Relative expression of Tet1, BDNF, NTF3, A20, FLIP, HSP70, HSP90, HSP27, and Bcl2 in neuron-like cells was detected by qPCR. In addition, neuron-like cells were divided into 7 groups. RESULTS Tet1 expression and deoxyribonucleic acid hydroxymethylation increased initially and decreased thereafter after neuron injury in both animal and cell models. Percentages of dead cells increased significantly in neuron-like cells after injury. The percentages of dead cells markedly decreased in injured neuron-like cells that were pretreated with azacytidine. The percentages of dead cells increased markedly in the Tet1 interference group and decreased significantly in the Tet1 overexpression group. Expression of Tet1, BDNF, A20, FLIP, HSP70, HSP90, and Bcl2 decreased significantly after injury. Azacytidine pretreatment in injured neuron-like cells markedly increased expression of Tet1, BDNF, NTF3, A20, FLIP, HSP70, HSP90, HSP27, and Bcl2. Moreover, Tet1 interference significantly decreased the expression of Tet1, BDNF, A20, FLIP, HSP70, and HSP90 in neuron-like cells, whereas Tet1 overexpression markedly increased the expression of Tet1, BDNF, NTF3, A20, FLIP, HSP70, HSP90, HSP27, and Bcl2. BDNF interference significantly increased percentages of dead cells after injury. BDNF interference also markedly decreased the protection of azacytidine and Tet1 overexpression against cell death. CONCLUSIONS Tet1 overexpression and demethylation caused by azacytidine protect neurons against cell death after injury by increasing expression of genes involved in cell survival.
Collapse
Affiliation(s)
- Yongxiang Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China.
| | - Jingcheng Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Hua Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Xinmin Feng
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Yuping Tao
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jiandong Yang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jun Cai
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou, China; Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
54
|
Ménard M, Costechareyre C, Coelho-Aguiar JM, Jarrosson-Wuilleme L, Rama N, Blachier J, Kindbeiter K, Bozon M, Cabrera JR, Dupin E, Le Douarin N, Mehlen P, Tauszig-Delamasure S. The dependence receptor TrkC regulates the number of sensory neurons during DRG development. Dev Biol 2018; 442:249-261. [DOI: 10.1016/j.ydbio.2018.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/17/2018] [Accepted: 07/26/2018] [Indexed: 11/28/2022]
|
55
|
Drug Targets in Neurotrophin Signaling in the Central and Peripheral Nervous System. Mol Neurobiol 2018; 55:6939-6955. [PMID: 29372544 DOI: 10.1007/s12035-018-0885-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/08/2018] [Indexed: 12/12/2022]
Abstract
Neurotrophins are a family of proteins that play an important role in the regulation of the growth, survival, and differentiation of neurons in the central and peripheral nervous system. Neurotrophins were earlier characterized by their role in early development, growth, maintenance, and the plasticity of the nervous system during development, but recent findings also indicate their complex role during normal physiology in both neuronal and non-neuronal tissues. Therefore, it is important to recognize a deficiency in the expression of neurotrophins, a major factor driving the debilitating features of several neurologic and psychiatric diseases/disorders. On the other hand, overexpression of neurotrophins is well known to play a critical role in pathogenesis of chronic pain and afferent sensitization, underlying conditions such as lower urinary tract symptoms (LUTS)/disorders and osteoarthritis. The existence of a redundant receptor system of high-and low-affinity receptors accounts for the diverse, often antagonistic, effects of neurotrophins in neurons and non-neuronal tissues in a spatial and temporal manner. In addition, studies looking at bladder dysfunction because of conditions such as spinal cord injury and diabetes mellitus have found alterations in the levels of these neurotrophins in the bladder, as well as in sensory afferent neurons, which further opens a new avenue for therapeutic targets. In this review, we will discuss the characteristics and roles of key neurotrophins and their involvement in the central and periphery nervous system in both normal and diseased conditions.
Collapse
|
56
|
Imai F, Yoshida Y. Molecular mechanisms underlying monosynaptic sensory-motor circuit development in the spinal cord. Dev Dyn 2018; 247:581-587. [PMID: 29226492 DOI: 10.1002/dvdy.24611] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023] Open
Abstract
Motor behaviors are precisely controlled by the integration of sensory and motor systems in the central nervous system (CNS). Proprioceptive sensory neurons, key components of the sensory system, are located in the dorsal root ganglia and project axons both centrally to the spinal cord and peripherally to muscles and tendons, communicating peripheral information about the body to the CNS. Changes in muscle length detected by muscle spindles, and tension variations in tendons conveyed by Golgi tendon organs, are communicated to the CNS through group Ia /II, and Ib proprioceptive sensory afferents, respectively. Group Ib proprioceptive sensory neurons connect with motor neurons indirectly through spinal interneurons, whereas group Ia/II axons form both direct (monosynaptic) and indirect connections with motor neurons. Although monosynaptic sensory-motor circuits between spindle proprioceptive sensory neurons and motor neurons have been extensively studied since 1950s, the molecular mechanisms underlying their formation and upkeep have only recently begun to be understood. We will discuss our current understanding of the molecular foundation of monosynaptic circuit development and maintenance involving proprioceptive sensory neurons and motor neurons in the mammalian spinal cord. Developmental Dynamics 247:581-587, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fumiyasu Imai
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yutaka Yoshida
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
57
|
Imai F, Chen X, Weirauch MT, Yoshida Y. Requirement for Dicer in Maintenance of Monosynaptic Sensory-Motor Circuits in the Spinal Cord. Cell Rep 2017; 17:2163-2172. [PMID: 27880894 DOI: 10.1016/j.celrep.2016.10.083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/08/2016] [Accepted: 10/24/2016] [Indexed: 01/19/2023] Open
Abstract
In contrast to our knowledge of mechanisms governing circuit formation, our understanding of how neural circuits are maintained is limited. Here, we show that Dicer, an RNaseIII protein required for processing microRNAs (miRNAs), is essential for maintenance of the spinal monosynaptic stretch reflex circuit in which group Ia proprioceptive sensory neurons form direct connections with motor neurons. In postnatal mice lacking Dicer in proprioceptor sensory neurons, there are no obvious defects in specificity or formation of monosynaptic sensory-motor connections. However, these circuits degrade through synapse loss and retraction of proprioceptive axonal projections from the ventral spinal cord. Peripheral terminals are also impaired without retracting from muscle targets. Interestingly, despite these central and peripheral axonal defects, proprioceptive neurons survive in the absence of Dicer-processed miRNAs. These findings reveal that Dicer, through its production of mature miRNAs, plays a key role in the maintenance of monosynaptic sensory-motor circuits.
Collapse
Affiliation(s)
- Fumiyasu Imai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Matthew T Weirauch
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
58
|
A RET-ER81-NRG1 Signaling Pathway Drives the Development of Pacinian Corpuscles. J Neurosci 2017; 36:10337-10355. [PMID: 27707970 DOI: 10.1523/jneurosci.2160-16.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/22/2016] [Indexed: 11/21/2022] Open
Abstract
Axon-Schwann cell interactions are crucial for the development, function, and repair of the peripheral nervous system, but mechanisms underlying communication between axons and nonmyelinating Schwann cells are unclear. Here, we show that ER81 is functionally required in a subset of mouse RET+ mechanosensory neurons for formation of Pacinian corpuscles, which are composed of a single myelinated axon and multiple layers of nonmyelinating Schwann cells, and Ret is required for the maintenance of Er81 expression. Interestingly, Er81 mutants have normal myelination but exhibit deficient interactions between axons and corpuscle-forming nonmyelinating Schwann cells. Finally, ablating Neuregulin-1 (Nrg1) in mechanosensory neurons results in no Pacinian corpuscles, and an Nrg1 isoform not required for communication with myelinating Schwann cells is specifically decreased in Er81-null somatosensory neurons. Collectively, our results suggest that a RET-ER81-NRG1 signaling pathway promotes axon communication with nonmyelinating Schwann cells, and that neurons use distinct mechanisms to interact with different types of Schwann cells. SIGNIFICANCE STATEMENT Communication between neurons and Schwann cells is critical for development, normal function, and regeneration of the peripheral nervous system. Despite many studies about axonal communication with myelinating Schwann cells, mostly via a specific isoform of Neuregulin1, the molecular nature of axonal communication with nonmyelinating Schwann cells is poorly understood. Here, we described a RET-ER81-Neuregulin1 signaling pathway in neurons innervating Pacinian corpuscle somatosensory end organs, which is essential for communication between the innervating axon and the end organ nonmyelinating Schwann cells. We also showed that this signaling pathway uses isoforms of Neuregulin1 that are not involved in myelination, providing evidence that neurons use different isoforms of Neuregulin1 to interact with different types of Schwann cells.
Collapse
|
59
|
Tail Nerve Electrical Stimulation and Electro-Acupuncture Can Protect Spinal Motor Neurons and Alleviate Muscle Atrophy after Spinal Cord Transection in Rats. Neural Plast 2017; 2017:7351238. [PMID: 28744378 PMCID: PMC5506460 DOI: 10.1155/2017/7351238] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 03/08/2017] [Accepted: 03/19/2017] [Indexed: 01/12/2023] Open
Abstract
Spinal cord injury (SCI) often results in death of spinal neurons and atrophy of muscles which they govern. Thus, following SCI, reorganizing the lumbar spinal sensorimotor pathways is crucial to alleviate muscle atrophy. Tail nerve electrical stimulation (TANES) has been shown to activate the central pattern generator (CPG) and improve the locomotion recovery of spinal contused rats. Electroacupuncture (EA) is a traditional Chinese medical practice which has been proven to have a neural protective effect. Here, we examined the effects of TANES and EA on lumbar motor neurons and hindlimb muscle in spinal transected rats, respectively. From the third day postsurgery, rats in the TANES group were treated 5 times a week and those in the EA group were treated once every other day. Four weeks later, both TANES and EA showed a significant impact in promoting survival of lumbar motor neurons and expression of choline acetyltransferase (ChAT) and ameliorating atrophy of hindlimb muscle after SCI. Meanwhile, the expression of neurotrophin-3 (NT-3) in the same spinal cord segment was significantly increased. These findings suggest that TANES and EA can augment the expression of NT-3 in the lumbar spinal cord that appears to protect the motor neurons as well as alleviate muscle atrophy.
Collapse
|
60
|
Nakahara Y, Gage FH, Tuszynski MH. Grafts of Fibroblasts Genetically Modified to Secrete Ngf, Bdnf, Nt-3, or Basic Fgf Elicit Differential Responses in the Adult Spinal Cord. Cell Transplant 2017; 5:191-204. [PMID: 8689031 DOI: 10.1177/096368979600500209] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Neuronal and axonal responses to neurotrophic factors in the developing spinal cord have been relatively well characterized, but little is known about adult spinal responses to neurotrophic factors. We genetically modified primary rat fibroblasts to produce either nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), or basic fibroblast growth factor (bFGF), then grafted these neurotrophic factor-secreting cells into the central gray matter of the spinal cord in adult rats. Spinal cord lesions were not made prior to grafting. From 2 wk to 6 mo later, sensory neurites of dorsal root origin extensively penetrated NGF-, NT-3-, and bFGF-producing grafts, whereas BDNF-secreting grafts elicited no growth responses. Putative noradrenergic neurites also penetrated NGF-secreting cell grafts. Local motor and corticospinal motor axons did not penetrate any of the neurotrophic factor-secreting grafts. These results indicate that unlesioned or minimally lesioned adult spinal cord sensory and putative noradrenergic populations retain significant neurotrophic factor responsiveness, whereas motor neurites are comparatively resistant even to those neurotrophic factors to which they exhibit survival dependence during development. Grafts of genetically modified cells can be a useful tool for characterizing neurotrophic factor responsiveness in the adult spinal cord and designing strategies to promote axonal regeneration after injury.
Collapse
Affiliation(s)
- Y Nakahara
- Department of Neurosciences, University of California-San Diego, La Jolla 92093, USA
| | | | | |
Collapse
|
61
|
Targeting Neurotrophins to Specific Populations of Neurons: NGF, BDNF, and NT-3 and Their Relevance for Treatment of Spinal Cord Injury. Int J Mol Sci 2017; 18:ijms18030548. [PMID: 28273811 PMCID: PMC5372564 DOI: 10.3390/ijms18030548] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Neurotrophins are a family of proteins that regulate neuronal survival, synaptic function, and neurotransmitter release, and elicit the plasticity and growth of axons within the adult central and peripheral nervous system. Since the 1950s, these factors have been extensively studied in traumatic injury models. Here we review several members of the classical family of neurotrophins, the receptors they bind to, and their contribution to axonal regeneration and sprouting of sensory and motor pathways after spinal cord injury (SCI). We focus on nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3), and their effects on populations of neurons within diverse spinal tracts. Understanding the cellular targets of neurotrophins and the responsiveness of specific neuronal populations will allow for the most efficient treatment strategies in the injured spinal cord.
Collapse
|
62
|
O'Toole SM, Ferrer MM, Mekonnen J, Zhang H, Shima Y, Ladle DR, Nelson SB. Dicer maintains the identity and function of proprioceptive sensory neurons. J Neurophysiol 2016; 117:1057-1069. [PMID: 28003412 DOI: 10.1152/jn.00763.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 11/22/2022] Open
Abstract
Neuronal cell identity is established during development and must be maintained throughout an animal's life (Fishell G, Heintz N. Neuron 80: 602-612, 2013). Transcription factors critical for establishing neuronal identity can be required for maintaining it (Deneris ES, Hobert O. Nat Neurosci 17: 899-907, 2014). Posttranscriptional regulation also plays an important role in neuronal differentiation (Bian S, Sun T. Mol Neurobiol 44: 359-373, 2011), but its role in maintaining cell identity is less established. To better understand how posttranscriptional regulation might contribute to cell identity, we examined the proprioceptive neurons in the dorsal root ganglion (DRG), a highly specialized sensory neuron class, with well-established properties that distinguish them from other neurons in the ganglion. By conditionally ablating Dicer in mice, using parvalbumin (Pvalb)-driven Cre recombinase, we impaired posttranscriptional regulation in the proprioceptive sensory neuron population. Knockout (KO) animals display a progressive form of ataxia at the beginning of the fourth postnatal week that is accompanied by a cell death within the DRG. Before cell loss, expression profiling shows a reduction of proprioceptor specific genes and an increased expression of nonproprioceptive genes normally enriched in other ganglion neurons. Furthermore, although central connections of these neurons are intact, the peripheral connections to the muscle are functionally impaired. Posttranscriptional regulation is therefore necessary to retain the transcriptional identity and support functional specialization of the proprioceptive sensory neurons.NEW & NOTEWORTHY We have demonstrated that selectively impairing Dicer in parvalbumin-positive neurons, which include the proprioceptors, triggers behavioral changes, a lack of muscle connectivity, and a loss of transcriptional identity as observed through RNA sequencing. These results suggest that Dicer and, most likely by extension, microRNAs are crucially important for maintaining proprioception. Additionally, this study hints at the larger question of how neurons maintain their functional and molecular specificity.
Collapse
Affiliation(s)
- Sean M O'Toole
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts; and
| | - Monica M Ferrer
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts; and
| | - Jennifer Mekonnen
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts; and
| | - Haihan Zhang
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts; and
| | - Yasuyuki Shima
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts; and
| | - David R Ladle
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, Ohio
| | - Sacha B Nelson
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts; and
| |
Collapse
|
63
|
O'Donovan KJ. Intrinsic Axonal Growth and the Drive for Regeneration. Front Neurosci 2016; 10:486. [PMID: 27833527 PMCID: PMC5081384 DOI: 10.3389/fnins.2016.00486] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/10/2016] [Indexed: 02/01/2023] Open
Abstract
Following damage to the adult nervous system in conditions like stroke, spinal cord injury, or traumatic brain injury, many neurons die and most of the remaining spared neurons fail to regenerate. Injured neurons fail to regrow both because of the inhibitory milieu in which they reside as well as a loss of the intrinsic growth capacity of the neurons. If we are to develop effective therapeutic interventions that promote functional recovery for the devastating injuries described above, we must not only better understand the molecular mechanisms of developmental axonal growth in hopes of re-activating these pathways in the adult, but at the same time be aware that re-activation of adult axonal growth may proceed via distinct mechanisms. With this knowledge in hand, promoting adult regeneration of central nervous system neurons can become a more tractable and realistic therapeutic endeavor.
Collapse
Affiliation(s)
- Kevin J O'Donovan
- Department of Chemistry and Life Science, United States Military Academy West Point, NY, USA
| |
Collapse
|
64
|
Naito Y, Lee AK, Takahashi H. Emerging roles of the neurotrophin receptor TrkC in synapse organization. Neurosci Res 2016; 116:10-17. [PMID: 27697534 DOI: 10.1016/j.neures.2016.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 09/18/2016] [Accepted: 09/20/2016] [Indexed: 10/20/2022]
Abstract
Tropomyosin-receptor-kinase (Trk) receptors have been extensively studied for their roles in kinase-dependent signaling cascades in nervous system development. Synapse organization is coordinated by trans-synaptic interactions of various cell adhesion proteins, a representative example of which is the neurexin-neuroligin complex. Recently, a novel role for TrkC as a synapse organizing protein has been established. Post-synaptic TrkC binds to pre-synaptic type-IIa receptor-type protein tyrosine phosphatase sigma (PTPσ). TrkC-PTPσ specifically induces excitatory synapses in a kinase domain-independent manner. TrkC has distinct extracellular domains for PTPσ- and NT-3-binding and thus may bind both ligands simultaneously. Indeed, NT-3 enhances the TrkC-PTPσ interaction, thus facilitating synapse induction at the pre-synaptic side and increasing pre-synaptic vesicle recycling in a kinase-independent fashion. A crystal structure study has revealed the detailed structure of the TrkC-PTPσ complex as well as competitive modulation of TrkC-mediated synaptogenesis by heparan sulfate proteoglycans (HSPGs), which bind the same domain of TrkC as PTPσ. Thus, there is strong evidence supporting a role for the TrkC-PTPσ complex in mechanisms underlying the fine turning of neural connectivity. Furthermore, disruption of the TrkC-PTPσ complex may be the underlying cause of certain psychiatric disorders caused by mutations in the gene encoding TrkC (NTRK3), supporting its role in cognitive functions.
Collapse
Affiliation(s)
- Yusuke Naito
- Synapse Development and Plasticity, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Alfred Kihoon Lee
- Synapse Development and Plasticity, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 2B4, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec H3A 1A3, Canada.
| |
Collapse
|
65
|
Abstract
Neurotrophin molecules have been shown to play important roles in the survival of neurons during devel opment. Most early studies concentrated on the initially discovered factor—nerve growth factor. Recent work has demonstrated that nerve growth factor belongs to a family of neurotrophins that include brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5. These neurotrophins exert their action via high- affinity receptors known as trks ( trkA, trkB, and trkC). A major goal of present research is to identify the neuronal locus of different trks to permit inferences about the specificity of action of the different growth factors. However, recent evidence suggests complex relationships between neurotrophins and trk receptors. These issues are explored in the context of dorsal root ganglion cells and motor neurons. Functional studies of the effects of nerve growth factor on its target cells, the nociceptive afferents, illustrate that this neurotro phin plays a role in development and function, which goes well beyond its classical role in promoting the survival of neurons during the early phases of development. The Neuroscientist 1:26-34, 1995
Collapse
Affiliation(s)
- Lorne M. Mendell
- Department of Neurobiology and Behavior SUNY at Stony
Brook Stony Brook, New York
| |
Collapse
|
66
|
Thiry L, Lemieux M, D Laflamme O, Bretzner F. Role of DSCAM in the development of the spinal locomotor and sensorimotor circuits. J Neurophysiol 2015; 115:1338-54. [PMID: 26655819 DOI: 10.1152/jn.00557.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 12/06/2015] [Indexed: 11/22/2022] Open
Abstract
Locomotion is controlled by spinal circuits that generate rhythm and coordinate left-right and flexor-extensor motoneuronal activities. The outputs of motoneurons and spinal interneuronal circuits are shaped by sensory feedback, relaying peripheral signals that are critical to the locomotor and postural control. Several studies in invertebrates and vertebrates have argued that the Down syndrome cell adhesion molecule (DSCAM) would play an important role in the normal development of neural circuits through cell spacing and targeting, axonal and dendritic branching, and synapse establishment and maintenance. Although there is evidence that DSCAM is important for the normal development of neural circuits, little is known about its functional contribution to spinal motor circuits. We show here that adult DSCAM(2J) mutant mice, lacking DSCAM, exhibit a higher variability in their locomotor pattern and rhythm during treadmill locomotion. Retrograde tracing studies in neonatal isolated spinal cords show an increased number of spinal commissural interneurons, which likely contributes to reducing the left-right alternation and to increasing the flexor/swing duration during neonatal and adult locomotion. Moreover, our results argue that, by reducing the peripheral excitatory drive onto spinal motoneurons, the DSCAM mutation reduces or abolishes spinal reflexes in both neonatal isolated spinal cords and adult mice, thus likely impairing sensorimotor control. Collectively, our functional, electrophysiological, and anatomical studies suggest that the mammalian DSCAM protein is involved in the normal development of spinal locomotor and sensorimotor circuits.
Collapse
Affiliation(s)
- Louise Thiry
- Centre de Recherche du Centre Hospitalier, (CHU) de Québec-CHUL and Département de Psychiatrie et Neurosciences de l'Université Laval, Québec, Québec, Canada
| | - Maxime Lemieux
- Centre de Recherche du Centre Hospitalier, (CHU) de Québec-CHUL and Département de Psychiatrie et Neurosciences de l'Université Laval, Québec, Québec, Canada
| | - Olivier D Laflamme
- Centre de Recherche du Centre Hospitalier, (CHU) de Québec-CHUL and Département de Psychiatrie et Neurosciences de l'Université Laval, Québec, Québec, Canada
| | - Frédéric Bretzner
- Centre de Recherche du Centre Hospitalier, (CHU) de Québec-CHUL and Département de Psychiatrie et Neurosciences de l'Université Laval, Québec, Québec, Canada
| |
Collapse
|
67
|
Sajgo S, Ali S, Popescu O, Badea TC. Dynamic expression of transcription factor Brn3b during mouse cranial nerve development. J Comp Neurol 2015; 524:1033-61. [PMID: 26356988 DOI: 10.1002/cne.23890] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/18/2015] [Accepted: 08/31/2015] [Indexed: 01/23/2023]
Abstract
During development, transcription factor combinatorial codes define a large variety of morphologically and physiologically distinct neurons. Such a combinatorial code has been proposed for the differentiation of projection neurons of the somatic and visceral components of cranial nerves. It is possible that individual neuronal cell types are not specified by unique transcription factors but rather emerge through the intersection of their expression domains. Brn3a, Brn3b, and Brn3c, in combination with each other and/or transcription factors of other families, can define subgroups of retinal ganglion cells (RGC), spiral and vestibular ganglia, inner ear and vestibular hair cell neurons in the vestibuloacoustic system, and groups of somatosensory neurons in the dorsal root ganglia. The present study investigates the expression and potential role of the Brn3b transcription factor in cranial nerves and associated nuclei of the brainstem. We report the dynamic expression of Brn3b in the somatosensory component of cranial nerves II, V, VII, and VIII and visceromotor nuclei of nerves VII, IX, and X as well as other brainstem nuclei during different stages of development into adult stage. We find that genetically identified Brn3b(KO) RGC axons show correct but delayed pathfinding during the early stages of embryonic development. However, loss of Brn3b does not affect the anatomy of the other cranial nerves normally expressing this transcription factor.
Collapse
Affiliation(s)
- Szilard Sajgo
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892.,Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Science, Babes-Bolyai University, Cluj-Napoca, Cluj, 400084, Romania
| | - Seid Ali
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892
| | - Octavian Popescu
- Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Science, Babes-Bolyai University, Cluj-Napoca, Cluj, 400084, Romania.,Institute of Biology, Romanian Academy, Bucharest, 060031, Romania
| | | |
Collapse
|
68
|
Abstract
Control of movement is a fundamental and complex task of the vertebrate nervous system, which relies on communication between circuits distributed throughout the brain and spinal cord. Many of the networks essential for the execution of basic locomotor behaviors are composed of discrete neuronal populations residing within the spinal cord. The organization and connectivity of these circuits is established through programs that generate functionally diverse neuronal subtypes, each contributing to a specific facet of motor output. Significant progress has been made in deciphering how neuronal subtypes are specified and in delineating the guidance and synaptic specificity determinants at the core of motor circuit assembly. Recent studies have shed light on the basic principles linking locomotor circuit connectivity with function, and they are beginning to reveal how more sophisticated motor behaviors are encoded. In this review, we discuss the impact of developmental programs in specifying motor behaviors governed by spinal circuits.
Collapse
Affiliation(s)
- Catarina Catela
- Neuroscience Institute and Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016;
| | - Maggie M Shin
- Neuroscience Institute and Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016;
| | - Jeremy S Dasen
- Neuroscience Institute and Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016;
| |
Collapse
|
69
|
Egr3-dependent muscle spindle stretch receptor intrafusal muscle fiber differentiation and fusimotor innervation homeostasis. J Neurosci 2015; 35:5566-78. [PMID: 25855173 DOI: 10.1523/jneurosci.0241-15.2015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Muscle stretch proprioceptors (muscle spindles) are required for stretch reflexes and locomotor control. Proprioception abnormalities are observed in many human neuropathies, but the mechanisms involved in establishing and maintaining muscle spindle innervation and function are still poorly understood. During skeletal muscle development, sensory (Ia-afferent) innervation induces contacted myotubes to transform into intrafusal muscle fibers that form the stretch receptor core. The transcriptional regulator Egr3 is induced in Ia-afferent contacted myotubes by Neuregulin1 (Nrg1)/ErbB receptor signaling and it has an essential role in spindle morphogenesis and function. Because Egr3 is widely expressed during development and has a pleiotropic function, whether Egr3 functions primarily in skeletal muscle, Ia-afferent neurons, or in Schwann cells that myelinate Ia-afferent axons remains unresolved. In the present studies, cell-specific ablation of Egr3 in mice showed that it has a skeletal muscle autonomous function in stretch receptor development. Moreover, using genetic tracing, we found that Ia-afferent contacted Egr3-deficient myotubes were induced in normal numbers, but their development was blocked to generate one to two shortened fibers that failed to express some characteristic myosin heavy chain (MyHC) proteins. These "spindle remnants" persisted into adulthood, remained innervated by Ia-afferents, and expressed neurotrophin3 (NT3), which is required for Ia-afferent neuron survival. However, they were not innervated by fusimotor axons and they did not express glial derived neurotrophic factor (GDNF), which is essential for fusimotor neuron survival. These results demonstrate that Egr3 has an essential role in regulating gene expression that promotes normal intrafusal muscle fiber differentiation and fusimotor innervation homeostasis.
Collapse
|
70
|
Embryonic neural stem cells in a 3D bioassay for trophic stimulation studies. Brain Res Bull 2015; 115:37-44. [DOI: 10.1016/j.brainresbull.2015.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 01/19/2023]
|
71
|
Abstract
Development of a functional peripheral nervous system requires axons to rapidly innervate and arborize into final target organs and then slow but not halt their growth to establish stable connections while keeping pace with organ growth. Here we examine the role of the NGF-TrkA effector protein, Coronin-1, on postganglionic sympathetic neuron final target innervation. In the absence of Coronin-1 we find that NGF-TrkA-PI3K signaling drives robust axon growth and branching in part by suppressing GSK3β. In contrast, the presence of Coronin-1 (wild-type neurons) suppresses but does not halt NGF-TrkA-dependent growth and branching. This relative suppression in axon growth behaviors is due to Coronin-1-dependent calcium release via PLC-γ1 signaling, which releases PI3K-dependent suppression of GSK3β. Finally, we demonstrate that Coro1a(-/-) mice display sympathetic axon overgrowth and overbranching phenotypes in the developing heart. Together with previous work demonstrating the Coronin-1 expression is NGF dependent, this work suggests that periods before and after NGF-TrkA-induced Coronin-1 expression (and likely other factors) defines two distinct axon growth states, which are critical for proper circuit formation in the sympathetic nervous system.
Collapse
|
72
|
Newbern JM. Molecular control of the neural crest and peripheral nervous system development. Curr Top Dev Biol 2015; 111:201-31. [PMID: 25662262 DOI: 10.1016/bs.ctdb.2014.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A transient and unique population of multipotent stem cells, known as neural crest cells (NCCs), generate a bewildering array of cell types during vertebrate development. An attractive model among developmental biologists, the study of NCC biology has provided a wealth of knowledge regarding the cellular and molecular mechanisms important for embryogenesis. Studies in numerous species have defined how distinct phases of NCC specification, proliferation, migration, and survival contribute to the formation of multiple functionally distinct organ systems. NCC contributions to the peripheral nervous system (PNS) are well known. Critical developmental processes have been defined that provide outstanding models for understanding how extracellular stimuli, cell-cell interactions, and transcriptional networks cooperate to direct cellular diversification and PNS morphogenesis. Dissecting the complex extracellular and intracellular mechanisms that mediate the formation of the PNS from NCCs may have important therapeutic implications for neurocristopathies, neuropathies, and certain forms of cancer.
Collapse
Affiliation(s)
- Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
73
|
Fantauzzo KA, Soriano P. Receptor tyrosine kinase signaling: regulating neural crest development one phosphate at a time. Curr Top Dev Biol 2015; 111:135-82. [PMID: 25662260 PMCID: PMC4363133 DOI: 10.1016/bs.ctdb.2014.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Receptor tyrosine kinases (RTKs) bind to a subset of growth factors on the surface of cells and elicit responses with broad roles in developmental and postnatal cellular processes. Receptors in this subclass consist of an extracellular ligand-binding domain, a single transmembrane domain, and an intracellular domain harboring a catalytic tyrosine kinase and regulatory sequences that are phosphorylated either by the receptor itself or by various interacting proteins. Once activated, RTKs bind signaling molecules and recruit effector proteins to mediate downstream cellular responses through various intracellular signaling pathways. In this chapter, we highlight the role of a subset of RTK families in regulating the activity of neural crest cells (NCCs) and the development of their derivatives in mammalian systems. NCCs are migratory, multipotent cells that can be subdivided into four axial populations, cranial, cardiac, vagal, and trunk. These cells migrate throughout the vertebrate embryo along defined pathways and give rise to unique cell types and structures. Interestingly, individual RTK families often have specific functions in a subpopulation of NCCs that contribute to the diversity of these cells and their derivatives in the mammalian embryo. We additionally discuss current methods used to investigate RTK signaling, including genetic, biochemical, large-scale proteomic, and biosensor approaches, which can be applied to study intracellular signaling pathways active downstream of this receptor subclass during NCC development.
Collapse
Affiliation(s)
- Katherine A Fantauzzo
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
74
|
Coles CH, Mitakidis N, Zhang P, Elegheert J, Lu W, Stoker AW, Nakagawa T, Craig AM, Jones EY, Aricescu AR. Structural basis for extracellular cis and trans RPTPσ signal competition in synaptogenesis. Nat Commun 2014; 5:5209. [PMID: 25385546 PMCID: PMC4239663 DOI: 10.1038/ncomms6209] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/09/2014] [Indexed: 01/26/2023] Open
Abstract
Receptor protein tyrosine phosphatase sigma (RPTPσ) regulates neuronal extension and acts as a presynaptic nexus for multiple protein and proteoglycan interactions during synaptogenesis. Unknown mechanisms govern the shift in RPTPσ function, from outgrowth promotion to synaptic organization. Here, we report crystallographic, electron microscopic and small-angle X-ray scattering analyses, which reveal sufficient inter-domain flexibility in the RPTPσ extracellular region for interaction with both cis (same cell) and trans (opposite cell) ligands. Crystal structures of RPTPσ bound to its postsynaptic ligand TrkC detail an interaction surface partially overlapping the glycosaminoglycan-binding site. Accordingly, heparan sulphate and heparin oligomers compete with TrkC for RPTPσ binding in vitro and disrupt TrkC-dependent synaptic differentiation in neuronal co-culture assays. We propose that transient RPTPσ ectodomain emergence from the presynaptic proteoglycan layer allows capture by TrkC to form a trans-synaptic complex, the consequent reduction in RPTPσ flexibility potentiating interactions with additional ligands to orchestrate excitatory synapse formation.
Collapse
Affiliation(s)
- Charlotte H. Coles
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Nikolaos Mitakidis
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Peng Zhang
- Brain Research Centre and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada V6T 2B5
| | - Jonathan Elegheert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Weixian Lu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Andrew W. Stoker
- Cancer Section, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, 702 Light Hall (0615), Nashville, Tennessee 37232-0615, USA
| | - Ann Marie Craig
- Brain Research Centre and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada V6T 2B5
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - A. Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
75
|
Chiang JH, Cheng WS, Hood L, Tian Q. An epigenetic biomarker panel for glioblastoma multiforme personalized medicine through DNA methylation analysis of human embryonic stem cell-like signature. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:310-23. [PMID: 24601786 DOI: 10.1089/omi.2013.0084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alterations of DNA methylation occur during the course of both stem cell development and tumorigenesis. We present a novel strategy that can be used to stratify glioblastoma multiforme (GBM) patients through the epigenetic states of genes associated with human embryonic stem cell (hESC) identity in order to 1) assess linkages between the methylation signatures of these stem cell genes and survival of GBM patients, and 2) delineate putative mechanisms leading to poor prognosis in some patient subgroups. A DNA methylation signature was established for stratifying GBM patients into several hESC methylator subgroups. The hESC methylator-negative phenotype has demonstrated poor survival and upregulation of glioma stem cell (GSC) markers, and is enriched in one of the previously defined transcriptomic phenotypes-the mesenchymal phenotype. We further identified a refined signature of 36 genes as the gene panel, including SOX2, POU3F2, FGFR2, GAP43, NTRK2, NTRK3, and NKX2-2, which are highly enriched in the nervous system. Both signatures outperformed the O6-methylguanine-DNA methyltransferase (MGMT) methylation test in predicting patient's outcome. These findings were also validated through an independent dataset of patients. Furthermore, through statistical analyses, both signatures were examined significantly. Hypomethylation of hESC-associated genes predicted poorer clinical outcome in GBM, supporting the idea that epigenetic activation of stem cell genes contributes to GBM aggression. The gene panel presented herein may be developed into clinical assays for patient stratification and future personalized medicine interventions.
Collapse
Affiliation(s)
- Jung-Hsien Chiang
- 1 Department of Computer Science and Information Engineering, National Cheng Kung University , Tainan City, Taiwan
| | | | | | | |
Collapse
|
76
|
Dekkers MPJ, Nikoletopoulou V, Barde YA. Cell biology in neuroscience: Death of developing neurons: new insights and implications for connectivity. ACTA ACUST UNITED AC 2014; 203:385-93. [PMID: 24217616 PMCID: PMC3824005 DOI: 10.1083/jcb.201306136] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The concept that target tissues determine the survival of neurons has inspired much of the thinking on neuronal development in vertebrates, not least because it is supported by decades of research on nerve growth factor (NGF) in the peripheral nervous system (PNS). Recent discoveries now help to understand why only some developing neurons selectively depend on NGF. They also indicate that the survival of most neurons in the central nervous system (CNS) is not simply regulated by single growth factors like in the PNS. Additionally, components of the cell death machinery have begun to be recognized as regulators of selective axonal degeneration and synaptic function, thus playing a critical role in wiring up the nervous system.
Collapse
|
77
|
Abstract
The tropomyosin-related tyrosine kinase (Trk) receptors were initially described as a family of growth factor receptors required for neuronal survival. They have since been shown to influence many aspects of neuronal development and function, including differentiation, outgrowth, and synaptic plasticity. This chapter will give an overview on the biology of Trk receptors within the nervous system. The structure and downstream signaling pathways of the full-length receptors will be described, as well as the biological functions of their truncated isoforms. Finally, the role of Trk receptors in the nervous system in health and disease will be discussed.
Collapse
|
78
|
Ceni C, Unsain N, Zeinieh MP, Barker PA. Neurotrophins in the regulation of cellular survival and death. Handb Exp Pharmacol 2014; 220:193-221. [PMID: 24668474 DOI: 10.1007/978-3-642-45106-5_8] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The neurotrophins play crucial roles regulating survival and apoptosis in the developing and injured nervous system. The four neurotrophins exert profound and crucial survival effects on developing peripheral neurons, and their expression and action is intimately tied to successful innervation of peripheral targets. In the central nervous system, they are dispensable for neuronal survival during development but support neuronal survival after lesion or other forms of injury. Neurotrophins also regulate apoptosis of both peripheral and central neurons, and we now recognize that there are regulatory advantages to having the same molecules regulate life and death decisions. This chapter examines the biological contexts in which these events take place and highlights the specific ligands, receptors, and signaling mechanisms that allow them to occur.
Collapse
Affiliation(s)
- Claire Ceni
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC, Canada, H3A 2B4
| | | | | | | |
Collapse
|
79
|
Neurotrophin signalling and transcription programmes interactions in the development of somatosensory neurons. Handb Exp Pharmacol 2014; 220:329-53. [PMID: 24668479 DOI: 10.1007/978-3-642-45106-5_13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Somatosensory neurons of the dorsal root ganglia are generated from multipotent neural crest cells by a process of progressive specification and differentiation. Intrinsic transcription programmes active in somatosensory neuron progenitors and early post-mitotic neurons drive the cell-type expression of neurotrophin receptors. In turn, signalling by members of the neurotrophin family controls expression of transcription factors that regulate neuronal sub-type specification. This chapter explores the mechanisms by which this crosstalk between neurotrophin signalling and transcription programmes generates the diverse functional sub-types of somatosensory neurons found in the mature animal.
Collapse
|
80
|
Yuan Y, Pan S, Sun Z, Dan Q, Liu J. Brain-derived neurotrophic factor-modified umbilical cord mesenchymal stem cell transplantation improves neurological deficits in rats with traumatic brain injury. Int J Neurosci 2013; 124:524-31. [DOI: 10.3109/00207454.2013.859144] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
81
|
Herndon CA, Ankenbruck N, Fromm L. The Erk MAP kinase pathway is activated at muscle spindles and is required for induction of the muscle spindle-specific gene Egr3 by neuregulin1. J Neurosci Res 2013; 92:174-84. [PMID: 24272970 DOI: 10.1002/jnr.23293] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 08/02/2013] [Accepted: 08/09/2013] [Indexed: 01/03/2023]
Abstract
Muscle spindles are sensory receptors composed of specialized muscle fibers, known as intrafusal muscle fibers, along with the endings of sensory neuron axons that innervate these muscle fibers. Formation of muscle spindles requires neuregulin1 (NRG1), which is released by sensory axons, activating ErbB receptors in muscle cells that are contacted. The transcription factor Egr3 is transcriptionally induced by NRG1, which in turn activates various target genes involved in forming intrafusal fibers. We have previously shown that, in cultured muscle cells, NRG1 signaling activates the Egr3 gene through SRF and CREB, which bind to a composite regulatory element, and that NRG1 signaling targets SRF by stimulating nuclear translocation of SRF coactivators myocardin-related transcription factor (MRTF)-A and MRTF-B and targets CREB by phosphorylation. The current studies examined signaling relays that might function in the NRG1 pathway upstream of SRF and CREB. We found that transcriptional induction of Egr3 in response to NRG1 requires the MAP kinase Erk1/2, which acts upstream of CREB to induce its phosphorylation. MRTFs are targeted by the Rho-actin pathway, yet in the absence of Rho-actin signaling, even though MRTFs fail to be translocated to the nucleus, NRG1 induces Egr3 transcription. In mouse muscle in vivo, activation of Erk1/2 is enhanced selectively where muscle spindles are located. These results suggest that Erk1/2 acts in intrafusal fibers of muscle spindles to induce transcription of Egr3 and that Egr3 induction occurs independently of MRTFs and involves Erk1/2 acting on other transcriptional regulatory targets that interact with the SRF-CREB regulatory element.
Collapse
Affiliation(s)
- Carter A Herndon
- Indiana University School of Medicine-Muncie and Ball State University, Muncie, Indiana
| | | | | |
Collapse
|
82
|
Fukuhara K, Imai F, Ladle DR, Katayama KI, Leslie JR, Arber S, Jessell TM, Yoshida Y. Specificity of monosynaptic sensory-motor connections imposed by repellent Sema3E-PlexinD1 signaling. Cell Rep 2013; 5:748-58. [PMID: 24210822 PMCID: PMC3844154 DOI: 10.1016/j.celrep.2013.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 08/04/2013] [Accepted: 10/02/2013] [Indexed: 11/23/2022] Open
Abstract
In mammalian spinal cord, group Ia proprioceptive afferents form selective monosynaptic connections with a select group of motor pool targets. The extent to which sensory recognition of motor neurons contributes to the selectivity of sensory-motor connections remains unclear. We show here that proprioceptive sensory afferents that express PlexinD1 avoid forming monosynaptic connections with neurons in Sema3E(+) motor pools yet are able to form direct connections with neurons in Sema3E(off) motor pools. Anatomical and electrophysiological analysis of mice in which Sema3E-PlexinD1 signaling has been deregulated or inactivated genetically reveals that repellent signaling underlies aspects of the specificity of monosynaptic sensory-motor connectivity in these reflex arcs. A semaphorin-based system of motor neuron recognition and repulsion therefore contributes to the formation of specific sensory-motor connections in mammalian spinal cord.
Collapse
Affiliation(s)
- Kaori Fukuhara
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Bartkowska K, Gajerska M, Turlejski K, Djavadian RL. Expression of TrkC receptors in the developing brain of the Monodelphis opossum and its effect on the development of cortical cells. PLoS One 2013; 8:e74346. [PMID: 24019963 PMCID: PMC3760877 DOI: 10.1371/journal.pone.0074346] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 07/31/2013] [Indexed: 02/05/2023] Open
Abstract
In this study, we investigated the distribution, localization and several various functions of TrkC receptors during development of the Monodelphisopossum brain. Western blotting analysis showed that two different forms of the TrkC receptor, the full-length receptor and one of its truncated forms, are abundantly expressed in the opossum brain. The expression of TrkC receptors was barely detected in the brain of newborn opossums. At postnatal day (P) 3, the expression of full-length TrkC remained at low levels, while moderate expression of the TrkC truncated form was detected. The expression levels of both forms of this protein gradually increased throughout development, peaking at P35. We found that in different neocortical areas located both at the rostral and caudal regions of the cortex, up to 98% of BrdU-labeled cells forming cortical layers (II-VI) had prominently expressed TrkC. To assess which developmental processes of cortical cells are regulated by TrkC receptors, three different shRNAs were constructed. The shRNAs were individually tested in transfected cortical progenitor cells grown on culture plates for 2 days. The effects of the shRNA-TrkC constructs were similar: blockade of TrkC receptors decreased the number of Ki67-positive and apoptotic cells, and it did not change the number of TUJ-positive neurons in vitro. Thus, the lack of TrkC receptors in cultured progenitor cells provided insight on the potential role of these receptors in the regulation of proliferation and cell survival but not in the differentiation of cortical cells.
Collapse
Affiliation(s)
- Katarzyna Bartkowska
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Monika Gajerska
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Kris Turlejski
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Rouzanna L. Djavadian
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
84
|
Pan Y, Zhang J, Liu W, Shu P, Yin B, Yuan J, Qiang B, Peng X. Dok5 is involved in the signaling pathway of neurotrophin-3 against TrkC-induced apoptosis. Neurosci Lett 2013; 553:46-51. [PMID: 23954828 DOI: 10.1016/j.neulet.2013.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/29/2013] [Accepted: 08/05/2013] [Indexed: 01/19/2023]
Abstract
TrkC is a dependence receptor and many reports have shown that neurotrophin-3 promotes cell survival by inhibiting TrkC-induced apoptosis in many cell lines. However, the identity of the adaptor protein involved in the NT-3/TrkC signaling pathway regulating cell death and survival remains unclear. The downstream of tyrosine kinase/docking protein (Dok) adaptor protein 5 is one substrate of the TrkC receptor. Because NT-3 and its receptor, TrkC, are strongly expressed by sensory neurons, we measured the expression of Dok5 and TrkC in the developing mouse spinal cord and dorsal root ganglia (DRG). We found that the number of cells positive for both Dok5 and TrkC decreases with DRG development. Immunoprecipitation and immunofluorescence staining showed that Dok5 interacted with TrkC and partially colocalized with TrkC in DRG neurons. In HEK293T cells, TrkC triggered apoptosis, but NT-3 prevented TrkC-induced apoptosis. Interestingly, siRNA knockdown of Dok5 expression partially prevented the protection of NT-3 against TrkC-induced apoptosis by regulating the activity of caspase-3. Taken together, we concluded that Dok5 is necessary for NT-3 signaling to block TrkC-induced apoptosis.
Collapse
Affiliation(s)
- Yanfang Pan
- The State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Cheret C, Willem M, Fricker FR, Wende H, Wulf-Goldenberg A, Tahirovic S, Nave KA, Saftig P, Haass C, Garratt AN, Bennett DL, Birchmeier C. Bace1 and Neuregulin-1 cooperate to control formation and maintenance of muscle spindles. EMBO J 2013; 32:2015-28. [PMID: 23792428 PMCID: PMC3715864 DOI: 10.1038/emboj.2013.146] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/29/2013] [Indexed: 01/18/2023] Open
Abstract
The protease β-secretase 1 (Bace1) was identified through its critical role in production of amyloid-β peptides (Aβ), the major component of amyloid plaques in Alzheimer's disease. Bace1 is considered a promising target for the treatment of this pathology, but processes additional substrates, among them Neuregulin-1 (Nrg1). Our biochemical analysis indicates that Bace1 processes the Ig-containing β1 Nrg1 (IgNrg1β1) isoform. We find that a graded reduction in IgNrg1 signal strength in vivo results in increasingly severe deficits in formation and maturation of muscle spindles, a proprioceptive organ critical for muscle coordination. Further, we show that Bace1 is required for formation and maturation of the muscle spindle. Finally, pharmacological inhibition and conditional mutagenesis in adult animals demonstrate that Bace1 and Nrg1 are essential to sustain muscle spindles and to maintain motor coordination. Our results assign to Bace1 a role in the control of coordinated movement through its regulation of muscle spindle physiology, and implicate IgNrg1-dependent processing as a molecular mechanism. Bace1 is required for Nrg1 processing for muscle spindle development. Bace1 inhibition leads to loss of motor coordination even in adult mice, suggesting potentially serious side effects for drugs targeting Bace1 as a treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Cyril Cheret
- Entwicklungsbiologie/Signaltransduktion, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
SAD kinases sculpt axonal arbors of sensory neurons through long- and short-term responses to neurotrophin signals. Neuron 2013; 79:39-53. [PMID: 23790753 DOI: 10.1016/j.neuron.2013.05.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2013] [Indexed: 11/20/2022]
Abstract
Extrinsic cues activate intrinsic signaling mechanisms to pattern neuronal shape and connectivity. We showed previously that three cytoplasmic Ser/Thr kinases, LKB1, SAD-A, and SAD-B, control early axon-dendrite polarization in forebrain neurons. Here, we assess their role in other neuronal types. We found that all three kinases are dispensable for axon formation outside of the cortex but that SAD kinases are required for formation of central axonal arbors by subsets of sensory neurons. The requirement for SAD kinases is most prominent in NT-3 dependent neurons. SAD kinases transduce NT-3 signals in two ways through distinct pathways. First, sustained NT-3/TrkC signaling increases SAD protein levels. Second, short-duration NT-3/TrkC signals transiently activate SADs by inducing dephosphorylation of C-terminal domains, thereby allowing activating phosphorylation of the kinase domain. We propose that SAD kinases integrate long- and short-duration signals from extrinsic cues to sculpt axon arbors within the CNS.
Collapse
|
87
|
Gajewska-Woźniak O, Skup M, Kasicki S, Ziemlińska E, Czarkowska-Bauch J. Enhancing proprioceptive input to motoneurons differentially affects expression of neurotrophin 3 and brain-derived neurotrophic factor in rat hoffmann-reflex circuitry. PLoS One 2013; 8:e65937. [PMID: 23776573 PMCID: PMC3679030 DOI: 10.1371/journal.pone.0065937] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/30/2013] [Indexed: 11/18/2022] Open
Abstract
The importance of neurotrophin 3 (NT-3) for motor control prompted us to ask the question whether direct electrical stimulation of low-threshold muscle afferents, strengthening the proprioceptive signaling, could effectively increase the endogenous pool of this neurotrophin and its receptor TrkC in the Hoffmann-reflex (H-reflex) circuitry. The effects were compared with those of brain-derived neurotrophic factor (BDNF) and its TrkB receptor. Continuous bursts of stimuli were delivered unilaterally for seven days, 80 min daily, by means of a cuff-electrode implanted over the tibial nerve in awake rats. The H-reflex was recorded in the soleus muscle to control the strength of stimulation. Stimulation aimed at activation of Ia fibers produced a strong increase of NT-3 protein, measured with ELISA, in the lumbar L3-6 segments of the spinal cord and in the soleus muscle. This stimulation exerted much weaker effect on BDNF protein level which slightly increased only in L3-6 segments of the spinal cord. Increased protein level of NT-3 and BDNF corresponded to the changes of NT-3 mRNA and BDNF mRNA expression in L3-6 segments but not in the soleus muscle. We disclosed tissue-specificity of TrkC mRNA and TrkB mRNA responses. In the spinal cord TrkC and TrkB transcripts tended to decrease, whereas in the soleus muscle TrkB mRNA decreased and TrkC mRNA expression strongly increased, suggesting that stimulation of Ia fibers leads to sensitization of the soleus muscle to NT-3 signaling. The possibility of increasing NT-3/TrkC signaling in the neuromuscular system, with minor effects on BDNF/TrkB signaling, by means of low-threshold electrical stimulation of peripheral nerves, which in humans might be applied in non-invasive way, offers an attractive therapeutic tool.
Collapse
Affiliation(s)
- Olga Gajewska-Woźniak
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Małgorzata Skup
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- * E-mail: (JC-B); (MS)
| | - Stefan Kasicki
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Ewelina Ziemlińska
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Julita Czarkowska-Bauch
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
- * E-mail: (JC-B); (MS)
| |
Collapse
|
88
|
de Nooij JC, Doobar S, Jessell TM. Etv1 inactivation reveals proprioceptor subclasses that reflect the level of NT3 expression in muscle targets. Neuron 2013; 77:1055-68. [PMID: 23522042 DOI: 10.1016/j.neuron.2013.01.015] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2013] [Indexed: 01/12/2023]
Abstract
The organization of spinal reflex circuits relies on the specification of distinct classes of proprioceptive sensory neurons (pSN), but the factors that drive such diversity remain unclear. We report here that pSNs supplying distinct skeletal muscles differ in their dependence on the ETS transcription factor Etv1 for their survival and differentiation. The status of Etv1-dependence is linked to the location of proprioceptor muscle targets: pSNs innervating hypaxial and axial muscles depend critically on Etv1 for survival, whereas those innervating certain limb muscles are resistant to Etv1 inactivation. The level of NT3 expression in individual muscles correlates with Etv1-dependence and the loss of pSNs triggered by Etv1 inactivation can be prevented by elevating the level of muscle-derived NT3-revealing a TrkC-activated Etv1-bypass pathway. Our findings support a model in which the specification of aspects of pSN subtype character is controlled by variation in the level of muscle NT3 expression and signaling.
Collapse
Affiliation(s)
- Joriene C de Nooij
- Department of Neuroscience, Howard Hughes Medical Institute, Kavli Institute for Brain Science, Columbia University, New York, NY 10032, USA
| | | | | |
Collapse
|
89
|
Mosconi T, Arends JJ, Jacquin MF. Null mutations of NT-3 and Bax affect trigeminal ganglion cell number but not brainstem barrelette pattern formation. Somatosens Mot Res 2013; 30:114-9. [PMID: 23614607 DOI: 10.3109/08990220.2013.775118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Trigeminal ganglion (TG) neurons innervate the grid-like array of whisker follicles on the face of the mouse. Central TG axons project to the trigeminal (V) brainstem nuclear complex, including the nucleus principalis (PrV) and the spinal subnucleus interpolaris (SpVi), where they innervate barrelettes that are organized in a pattern that recapitulates the whisker pattern on the face. Neurotrophin-3 (NT-3) supports a population of TG cells that supply slowly adapting mechanoreceptors in the whisker pad. We examined mice at embryonic day 17 (E17) and on the day of birth (P0) with null mutations of NT-3, Bax, a proapoptotic gene associated with naturally occurring cell death, and Bax/NT-3 double knockout (KO) mutants to determine if: (1) the number of TG cells would be reduced; (2) eliminating the Bax gene would rescue the NT-3-dependent neurons; and (3) the central projections of the rescued axons in the Bax/NT-3 double KO mice would fail to develop the barrelette patterns in the PrV and SpVi subnuclei. In mice at E17, NT-3(-/-) mutants had 65% fewer TG neurons than found in age-matched wild-type (WT) mice, and at P0, the number was reduced by 55% (p < 0.001 for both). Bax null mutant mice at E17 had 132% of the WT number of TG cells (p < 0.001), although the numbers returned to WT levels by P0. Bax/NT-3 double KO mice at E17 had TG cell numbers equal to those seen in WT, but the double KO failed to retain WT TG neuron numbers in P0 mice (39% fewer cells; p < 0.001). In all cases of reduced experimental neuron numbers, and in the E17 Bax(-/-) mice with supernumerary cells, the barrelette patterns in the PrV and SpVi were normal. Only a slight qualitative reduction in overall barrelette field area and clarity of barrelettes were seen. These results suggest that NT-3 is not necessary for barrelette pattern formation in the brainstem.
Collapse
Affiliation(s)
- Tony Mosconi
- Department of Physical Therapy, Western University of Health Sciences, Pomona, CA, USA
| | | | | |
Collapse
|
90
|
Herndon CA, Ankenbruck N, Lester B, Bailey J, Fromm L. Neuregulin1 signaling targets SRF and CREB and activates the muscle spindle-specific gene Egr3 through a composite SRF-CREB-binding site. Exp Cell Res 2013; 319:718-30. [PMID: 23318675 DOI: 10.1016/j.yexcr.2013.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/14/2012] [Accepted: 01/07/2013] [Indexed: 12/20/2022]
Abstract
Muscle spindles are sensory receptors embedded within muscle that detect changes in muscle length. Each spindle is composed of specialized muscle fibers, known as intrafusal muscle fibers, along with the endings of axons from sensory neurons that innervate these muscle fibers. Formation of muscle spindles requires neuregulin1 (NRG1), which is released by sensory axons, activating ErbB receptors in muscle cells that are contacted. In muscle cells, the transcription factor Egr3 is transcriptionally induced by NRG1, which in turn activates various target genes involved in forming the intrafusal fibers of muscle spindles. The signaling relay within the NRG1-ErbB pathway that acts to induce Egr3 is presumably critical for muscle spindle formation but for the most part has not been determined. In the current studies, we examined, using cultured muscle cells, transcriptional regulatory mechanisms by which Egr3 responds to NRG1. We identified a composite regulatory element for the Egr3 gene, consisting adjacent sites that bind cAMP response element binding protein (CREB) and serum response factor (SRF), with a role in NRG1 responsiveness. The SRF element also influences Egr3 basal expression in unstimulated myotubes, and in the absence of the SRF element, the CREB element influences basal expression. We show that NRG1 signaling, to target SRF, acts on the SRF coactivators myocardian-related transcription factor (MRTF)-A and MRTF-B, which are known to activate SRF-mediated transcription, by stimulating their translocation from the cytoplasm to the nucleus. CREB is phosphorylated, which is known to contribute to its activation, in response to NRG1. These results suggest that NRG1 induces expression of the muscle spindle-specific gene Egr3 by stimulating the transcriptional activity of CREB and SRF.
Collapse
Affiliation(s)
- Carter A Herndon
- Indiana University School of Medicine-Muncie and Ball State University, 2000 University Avenue, Muncie, IN 47306, USA
| | | | | | | | | |
Collapse
|
91
|
Hougland MT, Harrison BJ, Magnuson DSK, Rouchka EC, Petruska JC. The Transcriptional Response of Neurotrophins and Their Tyrosine Kinase Receptors in Lumbar Sensorimotor Circuits to Spinal Cord Contusion is Affected by Injury Severity and Survival Time. Front Physiol 2013; 3:478. [PMID: 23316162 PMCID: PMC3540763 DOI: 10.3389/fphys.2012.00478] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 12/07/2012] [Indexed: 01/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) results in changes to the anatomical, neurochemical, and physiological properties of cells in the central and peripheral nervous system. Neurotrophins, acting by binding to their cognate Trk receptors on target cell membranes, contribute to modulation of anatomical, neurochemical, and physiological properties of neurons in sensorimotor circuits in both the intact and injured spinal cord. Neurotrophin signaling is associated with many post-SCI changes including maladaptive plasticity leading to pain and autonomic dysreflexia, but also therapeutic approaches such as training-induced locomotor improvement. Here we characterize expression of mRNA for neurotrophins and Trk receptors in lumbar dorsal root ganglia (DRG) and spinal cord after two different severities of mid-thoracic injury and at 6 and 12 weeks post-SCI. There was complex regulation that differed with tissue, injury severity, and survival time, including reversals of regulation between 6 and 12 weeks, and the data suggest that natural regulation of neurotrophins in the spinal cord may continue for months after birth. Our assessments determined that a coordination of gene expression emerged at the 12-week post-SCI time point and bioinformatic analyses address possible mechanisms. These data can inform studies meant to determine the role of the neurotrophin signaling system in post-SCI function and plasticity, and studies using this signaling system as a therapeutic approach.
Collapse
Affiliation(s)
- M Tyler Hougland
- Department of Anatomical Sciences and Neurobiology, University of Louisville Louisville, KY, USA ; Laboratory of Neural Physiology and Plasticity, Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery Louisville, KY, USA
| | | | | | | | | |
Collapse
|
92
|
Lee J, Friese A, Mielich M, Sigrist M, Arber S. Scaling proprioceptor gene transcription by retrograde NT3 signaling. PLoS One 2012; 7:e45551. [PMID: 23029089 PMCID: PMC3447004 DOI: 10.1371/journal.pone.0045551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 08/23/2012] [Indexed: 01/12/2023] Open
Abstract
Cell-type specific intrinsic programs instruct neuronal subpopulations before target-derived factors influence later neuronal maturation. Retrograde neurotrophin signaling controls neuronal survival and maturation of dorsal root ganglion (DRG) sensory neurons, but how these potent signaling pathways intersect with transcriptional programs established at earlier developmental stages remains poorly understood. Here we determine the consequences of genetic alternation of NT3 signaling on genome-wide transcription programs in proprioceptors, an important sensory neuron subpopulation involved in motor reflex behavior. We find that the expression of many proprioceptor-enriched genes is dramatically altered by genetic NT3 elimination, independent of survival-related activities. Combinatorial analysis of gene expression profiles with proprioceptors isolated from mice expressing surplus muscular NT3 identifies an anticorrelated gene set with transcriptional levels scaled in opposite directions. Voluntary running experiments in adult mice further demonstrate the maintenance of transcriptional adjustability of genes expressed by DRG neurons, pointing to life-long gene expression plasticity in sensory neurons.
Collapse
Affiliation(s)
- Jun Lee
- Biozentrum, Department of Cell Biology, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Andreas Friese
- Biozentrum, Department of Cell Biology, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Monika Mielich
- Biozentrum, Department of Cell Biology, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Markus Sigrist
- Biozentrum, Department of Cell Biology, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Silvia Arber
- Biozentrum, Department of Cell Biology, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
93
|
Soren Leonard A, Puranam RS, Helgager J, Liu G, McNamara JO. Conditional deletion of TrkC does not modify limbic epileptogenesis. Epilepsy Res 2012; 102:126-30. [PMID: 22980123 DOI: 10.1016/j.eplepsyres.2012.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 07/16/2012] [Accepted: 07/30/2012] [Indexed: 02/01/2023]
Abstract
The neurotrophin receptor, tropomyosin-related kinase B (TrkB), is required for epileptogenesis in the kindling model. The role of a closely related neurotrophin receptor, TrkC, in limbic epileptogenesis is unknown. We examined limbic epileptogenesis in the kindling model in TrkC conditional null mice, using a strategy that previously established a critical role of TrkB. Despite elimination of TrkC mRNA, no differences in development of kindling were detected between TrkC conditional null and wild type control mice. These findings reinforce the central role of TrkB as the principal neurotrophin receptor involved in limbic epileptogenesis.
Collapse
Affiliation(s)
- A Soren Leonard
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
94
|
Kühne C, Puk O, Graw J, Hrabě de Angelis M, Schütz G, Wurst W, Deussing JM. Visualizing corticotropin-releasing hormone receptor type 1 expression and neuronal connectivities in the mouse using a novel multifunctional allele. J Comp Neurol 2012; 520:3150-80. [DOI: 10.1002/cne.23082] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
95
|
Pavan WJ, Raible DW. Specification of neural crest into sensory neuron and melanocyte lineages. Dev Biol 2012; 366:55-63. [PMID: 22465373 PMCID: PMC3351495 DOI: 10.1016/j.ydbio.2012.02.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/29/2012] [Indexed: 11/27/2022]
Abstract
Elucidating the mechanisms by which multipotent cells differentiate into distinct lineages is a common theme underlying developmental biology investigations. Progress has been made in understanding some of the essential factors and pathways involved in the specification of different lineages from the neural crest. These include gene regulatory networks involving transcription factor hierarchies and input from signaling pathways mediated from environmental cues. In this review, we examine the mechanisms for two lineages that are derived from the neural crest, peripheral sensory neurons and melanocytes. Insights into the specification of these cell types may reveal common themes in the specification processes that occur throughout development.
Collapse
Affiliation(s)
- William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
96
|
Guo X, Ayala JE, Gonzalez M, Stancescu M, Lambert S, Hickman JJ. Tissue engineering the monosynaptic circuit of the stretch reflex arc with co-culture of embryonic motoneurons and proprioceptive sensory neurons. Biomaterials 2012; 33:5723-31. [PMID: 22594977 DOI: 10.1016/j.biomaterials.2012.04.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 04/16/2012] [Indexed: 01/08/2023]
Abstract
The sensory circuit of the stretch reflex arc is composed of intrafusal muscle fibers and their innervating proprioceptive neurons that convert mechanical information regarding muscle length and tension into action potentials that synapse onto the homonymous motoneurons in the ventral spinal cord which innervate the extrafusal fibers of the same muscle. To date, the in vitro synaptic connection between proprioceptive sensory neurons and spinal motoneurons has not been demonstrated. A functional in vitro system demonstrating this connection would enable the understanding of feedback by the integration of sensory input into the spinal reflex arc. Here we report a co-culture of rat embryonic motoneurons and proprioceptive sensory neurons from dorsal root ganglia (DRG) in a defined serum-free medium on a synthetic silane substrate (DETA). Furthermore, we have demonstrated functional synapse formation in the co-culture by immunocytochemistry and electrophysiological analysis. This work will be valuable for enabling in vitro model systems for the study of spinal motor control and related pathologies such as spinal cord injury, muscular dystrophy and spasticity by improving our understanding of the integration of the mechanosensitive feedback mechanism.
Collapse
Affiliation(s)
- Xiufang Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | | | | | | | | | | |
Collapse
|
97
|
Development of primary sensory neurons in the trigeminal nervous system; dependency on neurotrophins and other substances. JAPANESE DENTAL SCIENCE REVIEW 2012. [DOI: 10.1016/j.jdsr.2011.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
98
|
Salemi M, La Vignera S, Castiglione R, Condorelli RA, Cimino L, Bosco P, Romano C, Romano C, Calogero AE. Expression of STRBP mRNA in patients with cryptorchidism and Down's syndrome. J Endocrinol Invest 2012; 35:5-7. [PMID: 22391137 DOI: 10.1007/bf03345414] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The most frequent defect of the male urogenital tract at birth is cryptorchidism. Cryptorchidism causes primitive testicular pathology responsible for infertility. Men with Down's syndrome (DS) have an increased risk of cryptorchidism. The spermatid perinuclear RNA-binding protein (STRBP) gene codifies a microtubule-associated RNA-binding protein and it is highly expressed in the testis as well as in the brain. At both levels, this gene seems to play a relevant role in the regular development of these organs. These observations prompted us to evaluate the expression of STRBP mRNA in 5 DS men with cryptorchidism and 5 normal healthy men (controls) by quantitative Real Time PCR in peripheral blood leukocytes. We found a decreased expression of the STRBP gene in men with DS and cryptorchidism compared with controls. This finding suggests that the impaired expression of this gene in DS may play a pathogenetic role in the altered brain and testicular development in subjects with DS and cryptorchidism.
Collapse
Affiliation(s)
- M Salemi
- Laboratory of Cytogenetics, Oasi Institute for Research on Mental Retardation and Brain Aging, Troina (EN), Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Smith GM, Falone AE, Frank E. Sensory axon regeneration: rebuilding functional connections in the spinal cord. Trends Neurosci 2011; 35:156-63. [PMID: 22137336 DOI: 10.1016/j.tins.2011.10.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 10/26/2011] [Accepted: 10/26/2011] [Indexed: 11/17/2022]
Abstract
Functional regeneration within the adult spinal cord remains a formidable task. A major barrier to regeneration of sensory axons into the spinal cord is the dorsal root entry zone. This region displays many of the inhibitory features characteristic of other central nervous system injuries. Several experimental treatments, including inactivation of inhibitory molecules (such as Nogo and chondroitin sulfate proteoglycans) or administration of neurotrophic factors (such as nerve growth factor, neurotrophin3, glial-derived neurotrophic factor and artemin), have been found to promote anatomical and functional regeneration across this barrier. However, there have been relatively few experiments to determine whether regenerating axons project back to their appropriate target areas within the spinal cord. This review focuses on recent advances in sensory axon regeneration, including studies assessing the ability of sensory axons to reconnect with their original synaptic targets.
Collapse
Affiliation(s)
- George M Smith
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536-0509, USA
| | | | | |
Collapse
|
100
|
Abstract
Mammalian skeletal muscle comprises different fiber types, whose identity is first established during embryonic development by intrinsic myogenic control mechanisms and is later modulated by neural and hormonal factors. The relative proportion of the different fiber types varies strikingly between species, and in humans shows significant variability between individuals. Myosin heavy chain isoforms, whose complete inventory and expression pattern are now available, provide a useful marker for fiber types, both for the four major forms present in trunk and limb muscles and the minor forms present in head and neck muscles. However, muscle fiber diversity involves all functional muscle cell compartments, including membrane excitation, excitation-contraction coupling, contractile machinery, cytoskeleton scaffold, and energy supply systems. Variations within each compartment are limited by the need of matching fiber type properties between different compartments. Nerve activity is a major control mechanism of the fiber type profile, and multiple signaling pathways are implicated in activity-dependent changes of muscle fibers. The characterization of these pathways is raising increasing interest in clinical medicine, given the potentially beneficial effects of muscle fiber type switching in the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Stefano Schiaffino
- Venetian Institute of Molecular Medicine, Department of Biomedical Sciences, University of Padova, Consiglio Nazionale delle Ricerche Institute of Neurosciences, and Department of Human Anatomy and Physiology, University of Padova, Padova, Italy
| | - Carlo Reggiani
- Venetian Institute of Molecular Medicine, Department of Biomedical Sciences, University of Padova, Consiglio Nazionale delle Ricerche Institute of Neurosciences, and Department of Human Anatomy and Physiology, University of Padova, Padova, Italy
| |
Collapse
|