Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 171(2): 440-455.e14, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28942925

ABSTRACT

Corticospinal neurons (CSNs) represent the direct cortical outputs to the spinal cord and play important roles in motor control across different species. However, their organizational principle remains unclear. By using a retrograde labeling system, we defined the requirement of CSNs in the execution of a skilled forelimb food-pellet retrieval task in mice. In vivo imaging of CSN activity during performance revealed the sequential activation of topographically ordered functional ensembles with moderate local mixing. Region-specific manipulations indicate that CSNs from caudal or rostral forelimb area control reaching or grasping, respectively, and both are required in the transitional pronation step. These region-specific CSNs terminate in different spinal levels and locations, therefore preferentially connecting with the premotor neurons of muscles engaged in different steps of the task. Together, our findings suggest that spatially defined groups of CSNs encode different movement modules, providing a logic for parallel-ordered corticospinal circuits to orchestrate multistep motor skills.


Subject(s)
Cervical Cord/physiology , Motor Skills , Neural Pathways , Animals , Calcium/analysis , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cervical Cord/cytology , Forelimb/physiology , Joints/physiology , Mice , Mice, Inbred C57BL
2.
EMBO J ; 36(9): 1134-1146, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28258061

ABSTRACT

Conventionally, neuronal development is regarded to follow a stereotypic sequence of neurogenesis, migration, and differentiation. We demonstrate that this notion is not a general principle of neuronal development by documenting the timing of mitosis in relation to multiple differentiation events for bipolar cells (BCs) in the zebrafish retina using in vivo imaging. We found that BC progenitors undergo terminal neurogenic divisions while in markedly disparate stages of neuronal differentiation. Remarkably, the differentiation state of individual BC progenitors at mitosis is not arbitrary but matches the differentiation state of post-mitotic BCs in their surround. By experimentally shifting the relative timing of progenitor division and differentiation, we provide evidence that neurogenesis and differentiation can occur independently of each other. We propose that the uncoupling of neurogenesis and differentiation could provide neurogenic programs with flexibility, while allowing for synchronous neuronal development within a continuously expanding cell pool.


Subject(s)
Cell Differentiation , Cell Division , Neurogenesis , Retina/embryology , Retinal Bipolar Cells/physiology , Zebrafish/embryology , Animals
3.
Proc Natl Acad Sci U S A ; 110(37): 15109-14, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23980162

ABSTRACT

Proper functioning of sensory systems requires the generation of appropriate numbers and proportions of neuronal subtypes that encode distinct information. Perception of color relies on signals from multiple cone photoreceptor types. In cone-dominated retinas, each cone expresses a single opsin type with peak sensitivity to UV, long (L) (red), medium (M) (green), or short (S) (blue) wavelengths. The modes of cell division generating distinct cone types are unknown. We report here a mechanism whereby zebrafish cone photoreceptors of the same type are produced by symmetric division of dedicated precursors. Transgenic fish in which the thyroid hormone receptor ß2 (trß2) promoter drives fluorescent protein expression before L-cone precursors themselves are produced permitted tracking of their division in vivo. Every L cone in a local region resulted from the terminal division of an L-cone precursor, suggesting that such divisions contribute significantly to L-cone production. Analysis of the fate of isolated pairs of cones and time-lapse observations suggest that other cone types can also arise by symmetric terminal divisions. Such divisions of dedicated precursors may help to rapidly attain the final numbers and proportions of cone types (L > M, UV > S) in zebrafish larvae. Loss- and gain-of-function experiments show that L-opsin expression requires trß2 activity before cone differentiation. Ectopic expression of trß2 after cone differentiation produces cones with mixed opsins. Temporal differences in the onset of trß2 expression could explain why some species have mixed, and others have pure, cone types.


Subject(s)
Cone Opsins/metabolism , Retinal Cone Photoreceptor Cells/cytology , Zebrafish/growth & development , Animals , Animals, Genetically Modified , Base Sequence , Cell Differentiation , Cell Division , Cell Lineage , Cone Opsins/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Larva/cytology , Larva/growth & development , Larva/metabolism , Luminescent Proteins/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Recombinant Proteins/genetics , Retinal Cone Photoreceptor Cells/classification , Retinal Cone Photoreceptor Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Thyroid Hormone Receptors beta/antagonists & inhibitors , Thyroid Hormone Receptors beta/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
bioRxiv ; 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39071408

ABSTRACT

A Hebbian model of circuit remodeling predicts that two sets of inputs with sufficiently distinct activity patterns will synaptically capture separate sets of target cells. Mice in which a subset of retinal ganglion cells (RGCs) target the wrong region of the dorsal lateral geniculate nucleus (dLGN) provide the conditions for testing this prediction. In albino mice, mistargeted RGC axons form an island of terminals that is distinct from the surrounding neuropil. Blocking retinal activity during development prevents the formation of this island. However, the synaptic connectivity of the island was unknown. Here, we combine light and electron microscopy to determine if this activity-dependent island of axon terminals represent a synaptically segregated subcircuit. We reconstructed the microcircuitry of the boundary between the island and non-island RGCs and found a remarkably strong segregation within retinogeniculate connectivity. We conclude that, when sets of retinal input are established in the wrong part of the dLGN, the developing circuitry responds by forming a synaptically isolated subcircuit from the otherwise fully connected network. The fact that there is a developmental starting condition that can induce a synaptically segregated microcircuit has important implications for our understanding of the organization of visual circuits and for our understanding of the implementation of activity dependent development.

5.
Cell Rep ; 42(10): 113165, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37751356

ABSTRACT

Retinal ganglion cell (RGC) degeneration drives vision loss in blinding conditions. RGC death is often triggered by axon degeneration in the optic nerve. Here, we study the contributions of dynamic and homeostatic Ca2+ levels to RGC death from axon injury. We find that axonal Ca2+ elevations from optic nerve injury do not propagate over distance or reach RGC somas, and acute and chronic Ca2+ dynamics do not affect RGC survival. Instead, we discover that baseline Ca2+ levels vary widely between RGCs and predict their survival after axon injury, and that lowering these levels reduces RGC survival. Further, we find that well-surviving RGC types have higher baseline Ca2+ levels than poorly surviving types. Finally, we observe considerable variation in the baseline Ca2+ levels of different RGCs of the same type, which are predictive of within-type differences in survival.


Subject(s)
Optic Nerve Injuries , Humans , Animals , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/metabolism , Calcium/metabolism , Axons/metabolism , Optic Nerve/metabolism , Cell Survival , Disease Models, Animal
6.
Neuron ; 56(4): 597-603, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18031679

ABSTRACT

Symmetric cell divisions have been proposed to rapidly increase neuronal number late in neurogenesis, but how critical this mode of division is to establishing a specific neuronal layer is unknown. Using in vivo time-lapse imaging methods, we discovered that in the laminated zebrafish retina, the horizontal cell (HC) layer forms quickly during embryonic development upon division of a precursor cell population. The precursor cells morphologically resemble immature, postmitotic HCs and express HC markers such as ptf1a and Prox1 prior to division. These precursors undergo nonapical symmetric division at the laminar location where mature HCs contact photoreceptors. Strikingly, the precursor cell type we observed generates exclusively HCs. We have thus identified a dedicated HC precursor, and our findings suggest a mechanism of neuronal layer formation whereby the location of mitosis could facilitate rapid contact between synaptic partners.


Subject(s)
Neural Pathways/cytology , Neural Pathways/embryology , Retina/embryology , Retinal Horizontal Cells/cytology , Retinal Horizontal Cells/embryology , Stem Cells/cytology , Animals , Biomarkers/metabolism , Body Patterning/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cell Shape/physiology , Green Fluorescent Proteins/genetics , Homeodomain Proteins/metabolism , Luminescent Proteins/genetics , Microscopy, Confocal , Neural Pathways/physiology , Organogenesis/physiology , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/physiology , Retina/cytology , Retinal Horizontal Cells/physiology , Stem Cells/physiology , Synapses/physiology , Synapses/ultrastructure , Time Factors , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Zebrafish
7.
J Neurosci ; 30(36): 11951-61, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826659

ABSTRACT

Astroglia secrete factors that promote synapse formation and maintenance. In culture, glial contact has also been shown to facilitate synaptogenesis. Here, we examined whether glial contact is important for establishing circuits in vivo by simultaneously monitoring differentiation of glial cells and local synaptogenesis over time. Photoreceptor circuits of the vertebrate retina are particularly suitable for this study because of the relatively simple, laminar organization of their connectivity with their target neurons, horizontal cells and bipolar cells. Also, individual photoreceptor terminals are ensheathed within the outer plexiform layer (OPL) by the processes of one type of glia, Müller glia cells (MGs). We conducted in vivo time-lapse multiphoton imaging of the rapidly developing and relatively transparent zebrafish retina to ascertain the time course of MG development relative to OPL synaptogenesis. The emergence of synaptic triads, indicative of functional photoreceptor circuits, and structural association with glial processes were also examined across ages by electron microscopy. We first show that MG processes form territories that tile within the inner and outer synaptic layers. We then demonstrate that cone photoreceptor synapses are assembled before the elaboration of MG processes in the OPL. Using a targeted cell ablation approach, we also determined whether the maintenance of photoreceptor synapses is perturbed when local MGs are absent. We found that removal of MGs had no appreciable effect on the stability of newly formed cone synapses. Thus, in contrast to other CNS circuits, contact from glia is not necessary for the formation or immediate stabilization of outer retinal synapses.


Subject(s)
Neuroglia/physiology , Neurons/physiology , Retina/cytology , Synapses/physiology , Amino Acids , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Imaging, Three-Dimensional/methods , Luminescent Proteins/genetics , Microscopy, Confocal/methods , Microscopy, Electron, Transmission/methods , Neuroglia/ultrastructure , Neurons/classification , Neurons/ultrastructure , Photobleaching , Receptors, Glutamate/metabolism , Synapses/ultrastructure , Time Factors , Zebrafish , Zebrafish Proteins/genetics
8.
J Environ Manage ; 92(8): 1996-2009, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21486685

ABSTRACT

Within the research field of urban water demand management, understanding the link between environmental and water conservation attitudes and observed end use water consumption has been limited. Through a mixed method research design incorporating field-based smart metering technology and questionnaire surveys, this paper reveals the relationship between environmental and water conservation attitudes and a domestic water end use break down for 132 detached households located in Gold Coast city, Australia. Using confirmatory factor analysis, attitudinal factors were developed and refined; households were then categorised based on these factors through cluster analysis technique. Results indicated that residents with very positive environmental and water conservation attitudes consumed significantly less water in total and across the behaviourally influenced end uses of shower, clothes washer, irrigation and tap, than those with moderately positive attitudinal concern. The paper concluded with implications for urban water demand management planning, policy and practice.


Subject(s)
Conservation of Natural Resources , Drinking , Economics/statistics & numerical data , Environment , Water Supply/statistics & numerical data , Cluster Analysis , Factor Analysis, Statistical , Health Knowledge, Attitudes, Practice , Humans , Public Opinion , Queensland
9.
J Vis Exp ; (168)2021 02 13.
Article in English | MEDLINE | ID: mdl-33645555

ABSTRACT

The retina transforms light signals from the environment into electrical signals that are propagated to the brain. Diseases of the retina are prevalent and cause visual impairment and blindness. Understanding how such diseases progress is critical to formulating new treatments. In vivo microscopy in animal models of disease is a powerful tool for understanding neurodegeneration and has led to important progress towards treatments of conditions ranging from Alzheimer's disease to stroke. Given that the retina is the only central nervous system structure inherently accessible by optical approaches, it naturally lends itself towards in vivo imaging. However, the native optics of the lens and cornea present some challenges for effective imaging access. This protocol outlines methods for in vivo two-photon imaging of cellular cohorts and structures in the mouse retina at cellular resolution, applicable for both acute- and chronic-duration imaging experiments. It presents examples of retinal ganglion cell (RGC), amacrine cell, microglial, and vascular imaging using a suite of labeling techniques including adeno-associated virus (AAV) vectors, transgenic mice, and inorganic dyes. Importantly, these techniques extend to all cell types of the retina, and suggested methods for accessing other cellular populations of interest are described. Also detailed are example strategies for manual image postprocessing for display and quantification. These techniques are directly applicable to studies of retinal function in health and disease.


Subject(s)
Photons , Pupil/physiology , Retina/diagnostic imaging , Animals , Calcium/metabolism , Dependovirus/metabolism , Image Processing, Computer-Assisted , Intravitreal Injections , Mice , Mice, Transgenic , Microglia/cytology , Retinal Ganglion Cells/cytology , Software
10.
Curr Biol ; 31(21): 4870-4878.e5, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34534440

ABSTRACT

Neuronal identity has long been thought of as immutable, so that once a cell acquires a specific fate, it is maintained for life.1 Studies using the overexpression of potent transcription factors to experimentally reprogram neuronal fate in the mouse neocortex2,3 and retina4,5 have challenged this notion by revealing that post-mitotic neurons can switch their identity. Whether fate reprogramming is part of normal development in the central nervous system (CNS) is unclear. While there are some reports of physiological cell fate reprogramming in invertebrates,6,7 and in the vertebrate peripheral nervous system,8 endogenous fate reprogramming in the vertebrate CNS has not been documented. Here, we demonstrate spontaneous fate re-specification in an interneuron lineage in the zebrafish retina. We show that the visual system homeobox 1 (vsx1)-expressing lineage, which has been associated exclusively with excitatory bipolar cell (BC) interneurons,9-12 also generates inhibitory amacrine cells (ACs). We identify a role for Notch signaling in conferring plasticity to nascent vsx1 BCs, allowing suitable transcription factor programs to re-specify them to an AC fate. Overstimulating Notch signaling enhances this physiological phenotype so that both daughters of a vsx1 progenitor differentiate into ACs and partially differentiated vsx1 BCs can be converted into ACs. Furthermore, this physiological re-specification can be mimicked to allow experimental induction of an entirely distinct fate, that of retinal projection neurons, from the vsx1 lineage. Our observations reveal unanticipated plasticity of cell fate during retinal development.


Subject(s)
Homeodomain Proteins , Zebrafish , Animals , Cell Differentiation/genetics , Cell Lineage , Central Nervous System , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mice , Neurons/physiology , Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
11.
Neuron ; 109(9): 1527-1539.e4, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33784498

ABSTRACT

Predators use vision to hunt, and hunting success is one of evolution's main selection pressures. However, how viewing strategies and visual systems are adapted to predation is unclear. Tracking predator-prey interactions of mice and crickets in 3D, we find that mice trace crickets with their binocular visual fields and that monocular mice are poor hunters. Mammalian binocular vision requires ipsi- and contralateral projections of retinal ganglion cells (RGCs) to the brain. Large-scale single-cell recordings and morphological reconstructions reveal that only a small subset (9 of 40+) of RGC types in the ventrotemporal mouse retina innervate ipsilateral brain areas (ipsi-RGCs). Selective ablation of ipsi-RGCs (<2% of RGCs) in the adult retina drastically reduces the hunting success of mice. Stimuli based on ethological observations indicate that five ipsi-RGC types reliably signal prey. Thus, viewing strategies align with a spatially restricted and cell-type-specific set of ipsi-RGCs that supports binocular vision to guide predation.


Subject(s)
Depth Perception/physiology , Predatory Behavior/physiology , Retinal Ganglion Cells , Vision, Binocular/physiology , Animals , Functional Laterality/physiology , Mice , Visual Pathways/cytology , Visual Pathways/physiology
12.
Neuron ; 52(4): 609-21, 2006 Nov 22.
Article in English | MEDLINE | ID: mdl-17114046

ABSTRACT

Targeting of axons and dendrites to particular synaptic laminae is an important mechanism by which precise patterns of neuronal connectivity are established. Although axons target specific laminae during development, dendritic lamination has been thought to occur largely by pruning of inappropriately placed arbors. We discovered by in vivo time-lapse imaging that retinal ganglion cell (RGC) dendrites in zebrafish show growth patterns implicating dendritic targeting as a mechanism for contacting appropriate synaptic partners. Populations of RGCs labeled in transgenic animals establish distinct dendritic strata sequentially, predominantly from the inner to outer retina. Imaging individual cells over successive days confirmed that multistratified RGCs generate strata sequentially, each arbor elaborating within a specific lamina. Simultaneous imaging of RGCs and subpopulations of presynaptic amacrine interneurons revealed that RGC dendrites appear to target amacrine plexuses that had already laminated. Dendritic targeting of prepatterned afferents may thus be a novel mechanism for establishing proper synaptic connectivity.


Subject(s)
Dendrites/ultrastructure , Presynaptic Terminals/ultrastructure , Retina/cytology , Retina/embryology , Retinal Ganglion Cells/cytology , Zebrafish/embryology , Afferent Pathways/cytology , Afferent Pathways/embryology , Afferent Pathways/physiology , Amacrine Cells/cytology , Amacrine Cells/physiology , Animals , Animals, Genetically Modified , Cell Communication/genetics , Cell Differentiation/physiology , Cell Shape/physiology , Dendrites/physiology , Gene Expression Regulation, Developmental/genetics , Image Cytometry , Luminescent Proteins/genetics , Microscopy, Confocal , Presynaptic Terminals/physiology , Retina/physiology , Retinal Ganglion Cells/physiology , Time Factors , Zebrafish/physiology
13.
Annu Rev Vis Sci ; 6: 195-213, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32936739

ABSTRACT

The damage or loss of retinal ganglion cells (RGCs) and their axons accounts for the visual functional defects observed after traumatic injury, in degenerative diseases such as glaucoma, or in compressive optic neuropathies such as from optic glioma. By using optic nerve crush injury models, recent studies have revealed the cellular and molecular logic behind the regenerative failure of injured RGC axons in adult mammals and suggested several strategies with translational potential. This review summarizes these findings and discusses challenges for developing clinically applicable neural repair strategies.


Subject(s)
Nerve Regeneration/physiology , Optic Nerve Injuries/physiopathology , Retinal Ganglion Cells/physiology , Animals , Cell Survival , Disease Models, Animal , Humans , Nerve Crush , Optic Nerve Diseases
14.
Neuron ; 103(1): 39-51.e5, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31122676

ABSTRACT

Despite robust effects on immature neurons, growth factors minimally promote axon regeneration in the adult central nervous system (CNS). Attempting to improve growth-factor responsiveness in mature neurons by dedifferentiation, we overexpressed Lin28 in the retina. Lin28-treated retinas responded to insulin-like growth factor-1 (IGF1) by initiating retinal ganglion cell (RGC) axon regeneration after axotomy. Surprisingly, this effect was cell non-autonomous. Lin28 expression was required only in amacrine cells, inhibitory neurons that innervate RGCs. Ultimately, we found that optic-nerve crush pathologically upregulated activity in amacrine cells, which reduced RGC electrical activity and suppressed growth-factor signaling. Silencing amacrine cells or pharmacologically blocking inhibitory neurotransmission also induced IGF1 competence. Remarkably, RGCs regenerating across these manipulations localized IGF1 receptor to their primary cilia, which maintained their signaling competence and regenerative ability. Thus, our results reveal a circuit-based mechanism that regulates CNS axon regeneration and implicate primary cilia as a regenerative signaling hub.


Subject(s)
Axons/physiology , Nerve Growth Factor/physiology , Nerve Regeneration/physiology , Receptors, Presynaptic/physiology , Amacrine Cells/physiology , Animals , Cilia/metabolism , Cilia/ultrastructure , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Nerve Crush , Optic Nerve Injuries/pathology , RNA-Binding Proteins/genetics , Receptor, IGF Type 1/metabolism , Retina/metabolism , Retinal Ganglion Cells/drug effects
15.
Neuron ; 101(4): 615-624.e5, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30686733

ABSTRACT

Axon loss determines persistent disability in multiple sclerosis patients. Here, we use in vivo calcium imaging in a multiple sclerosis model to show that cytoplasmic calcium levels determine the choice between axon loss and survival. We rule out the endoplasmic reticulum, glutamate excitotoxicity, and the reversal of the sodium-calcium exchanger as sources of intra-axonal calcium accumulation and instead identify nanoscale ruptures of the axonal plasma membrane as the critical path of calcium entry.


Subject(s)
Axons/metabolism , Calcium/metabolism , Cell Membrane/pathology , Multiple Sclerosis/metabolism , Animals , Axons/pathology , Cell Membrane/metabolism , Female , Ion Transport , Male , Mice , Multiple Sclerosis/etiology
16.
Neuron ; 95(4): 817-833.e4, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28817801

ABSTRACT

A major hurdle for functional recovery after both spinal cord injury and cortical stroke is the limited regrowth of the axons in the corticospinal tract (CST) that originate in the motor cortex and innervate the spinal cord. Despite recent advances in engaging the intrinsic mechanisms that control CST regrowth, it remains to be tested whether such methods can promote functional recovery in translatable settings. Here we show that post-lesional AAV-assisted co-expression of two soluble proteins, namely insulin-like growth factor 1 (IGF1) and osteopontin (OPN), in cortical neurons leads to robust CST regrowth and the recovery of CST-dependent behavioral performance after both T10 lateral spinal hemisection and a unilateral cortical stroke. In these mice, a compound able to increase axon conduction, 4-aminopyridine-3-methanol, promotes further improvement in CST-dependent behavioral tasks. Thus, our results demonstrate a potentially translatable strategy for restoring cortical dependent function after injury in the adult.


Subject(s)
Insulin-Like Growth Factor I/therapeutic use , Neurons/drug effects , Osteopontin/therapeutic use , Pyramidal Tracts/pathology , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Age Factors , Aminopyridines/pharmacology , Animals , Axotomy , Disease Models, Animal , Functional Laterality , Hindlimb/physiopathology , Insulin-Like Growth Factor I/pharmacology , Mice , Movement Disorders/drug therapy , Movement Disorders/etiology , Osteopontin/pharmacology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Recovery of Function/physiology , Reflex/drug effects , Reflex/genetics , Serotonin/metabolism , Stroke/drug therapy , Time Factors
17.
J Neurosci ; 24(39): 8562-76, 2004 Sep 29.
Article in English | MEDLINE | ID: mdl-15456830

ABSTRACT

Spinal cord injury (SCI) induces a progressive pathophysiology affecting cell survival and neurological integrity via complex and evolving molecular cascades whose interrelationships are not fully understood. The present experiments were designed to: (1) determine potential functional interactions within transcriptional expression profiles obtained after a clinically relevant SCI and (2) test the consistency of transcript expression after SCI in two genetically and immunologically diverse rat strains characterized by differences in T cell competence and associated inflammatory responses. By interrogating Affymetrix U34A rat genome GeneChip microarrays, we defined the transcriptional expression patterns in midcervical contusion lesion sites between 1 and 90 d postinjury of athymic nude (AN) and Sprague Dawley (SD) strains. Stringent statistical analyses detected significant changes in 3638 probe sets, with 80 genes differing between the AN and SD groups. Subsequent detailed functional categorization of these transcripts unveiled an overall tissue remodeling response that was common to both strains. The functionally organized gene profiles were temporally distinct and correlated with repair indices observed microscopically and by magnetic resonance microimaging. Our molecular and anatomical observations have identified a novel, longitudinal perspective of the post-SCI response, namely, that of a highly orchestrated tissue repair and remodeling repertoire with a prominent cutaneous wound healing signature that is conserved between two widely differing rat strains. These results have significant bearing on the continuing development of cellular and pharmacological therapeutics directed at tissue rescue and neuronal regeneration in the injured spinal cord.


Subject(s)
Nerve Tissue Proteins/biosynthesis , Spinal Cord Injuries/physiopathology , Wound Healing/physiology , Algorithms , Animals , Cell Hypoxia/physiology , Cell Movement , Cell Proliferation , Female , Gene Expression , Magnetic Resonance Imaging , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger , Rats , Rats, Nude , Rats, Sprague-Dawley , Skin/injuries , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , T-Lymphocytes/physiology , Time Factors , Wound Healing/genetics , Wound Healing/immunology
18.
Neuron ; 88(4): 704-19, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26526391

ABSTRACT

After axotomy, neuronal survival and growth cone re-formation are required for axon regeneration. We discovered that doublecortin-like kinases (DCLKs), members of the doublecortin (DCX) family expressed in adult retinal ganglion cells (RGCs), play critical roles in both processes, through distinct mechanisms. Overexpression of DCLK2 accelerated growth cone re-formation in vitro and enhanced the initiation and elongation of axon re-growth after optic nerve injury. These effects depended on both the microtubule (MT)-binding domain and the serine-proline-rich (S/P-rich) region of DCXs in-cis in the same molecules. While the MT-binding domain is known to stabilize MT structures, we show that the S/P-rich region prevents F-actin destabilization in injured axon stumps. Additionally, while DCXs synergize with mTOR to stimulate axon regeneration, alone they can promote neuronal survival possibly by regulating the retrograde propagation of injury signals. Multifunctional DCXs thus represent potential targets for promoting both survival and regeneration of injured neurons.


Subject(s)
Actins/metabolism , Axons/metabolism , Microtubules/metabolism , Nerve Regeneration/genetics , Protein Serine-Threonine Kinases/genetics , Retinal Ganglion Cells/metabolism , Animals , Axons/physiology , Axotomy , Cell Survival , Doublecortin Protein , Doublecortin-Like Kinases , Growth Cones , In Vitro Techniques , Mice , Nerve Regeneration/physiology , Neurons/metabolism , Neurons/physiology , Optic Nerve Injuries , Protein Serine-Threonine Kinases/metabolism , Retinal Ganglion Cells/physiology , TOR Serine-Threonine Kinases/metabolism
19.
Nat Commun ; 5: 5683, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25511170

ABSTRACT

Therapeutic strategies for spinal cord injury (SCI) commonly focus on regenerating disconnected axons. An alternative approach would be to maintain continuity of damaged axons, especially after contusion. The viability of such neuropreservative strategies depends on the degree to which initially injured axons can recover. Here we use morphological and molecular in vivo imaging after contusion SCI in mice to show that injured axons persist in a metastable state for hours. Intra-axonal calcium dynamics influence fate, but the outcome is not invariably destructive in that many axons with calcium elevations recover homeostasis without intervention. Calcium enters axons primarily through mechanopores. Spontaneous pore resealing allows calcium levels to normalize and axons to survive long term. Axon loss can be halted by blocking calcium influx or calpain, even with delayed initiation. Our data identify an inherent self-preservation process in contused axons and a window of opportunity for rescuing connectivity after nontransecting SCI.


Subject(s)
Recovery of Function/physiology , Spinal Cord Injuries/rehabilitation , Spinal Cord/ultrastructure , Animals , Axons/metabolism , Axons/ultrastructure , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium/metabolism , Calpain/metabolism , Cations, Divalent , Female , Gene Expression , Genes, Reporter , Ion Transport , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Molecular Imaging , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors
20.
Nat Commun ; 5: 3699, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24832361

ABSTRACT

Many neurons receive synapses in stereotypic proportions from converging but functionally distinct afferents. However, developmental mechanisms regulating synaptic convergence are not well understood. Here we describe a heterotypic mechanism by which one afferent controls synaptogenesis of another afferent, but not vice versa. Like other CNS circuits, zebrafish retinal H3 horizontal cells (HC) undergo an initial period of remodelling, establishing synapses with ultraviolet and blue cones while eliminating red and green cone contacts. As development progresses, the HCs selectively synapse with ultraviolet cones to generate a 5:1 ultraviolet-to-blue cone synapse ratio. Blue cone synaptogenesis increases in mutants lacking ultraviolet cones, and when transmitter release or visual stimulation of ultraviolet cones is perturbed. Connectivity is unaltered when blue cone transmission is suppressed. Moreover, there is no cell-autonomous regulation of cone synaptogenesis by neurotransmission. Thus, biased connectivity in this circuit is established by an unusual activity-dependent, unidirectional control of synaptogenesis exerted by the dominant input.


Subject(s)
Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synapses/physiology , Zebrafish , Animals , Neuronal Plasticity , Retina/physiology
SELECTION OF CITATIONS
SEARCH DETAIL