Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John L. Bixby is active.

Publication


Featured researches published by John L. Bixby.


Science | 2009

KLF Family Members Regulate Intrinsic Axon Regeneration Ability

Darcie L. Moore; Murray G. Blackmore; Ying Hu; Klaus H. Kaestner; John L. Bixby; Vance Lemmon; Jeffrey L. Goldberg

Containing Neuronal Exuberance In rats and mice, around the time of birth, neurons of the central nervous system switch from a growth mode and lose their ability to regenerate. Studying retinal ganglion cells of the rat, Moore et al. (p. 298; see the Perspective by Subang and Richardson) identified a gene, Krüppel-like factor-4 (KLF4), that seems to contribute to the switch. The KLF4 gene belongs to a family of related transcription factors that possess repressive or enhancing effects on axon growth. The combinatorial effect of this family of transcription factors before and after birth may fine-tune the ability of the neurons to extend axons. The regenerative capacity of mouse retinal ganglion cells after injury is regulated by the KLF family of transcription factors. Neurons in the central nervous system (CNS) lose their ability to regenerate early in development, but the underlying mechanisms are unknown. By screening genes developmentally regulated in retinal ganglion cells (RGCs), we identified Krüppel-like factor–4 (KLF4) as a transcriptional repressor of axon growth in RGCs and other CNS neurons. RGCs lacking KLF4 showed increased axon growth both in vitro and after optic nerve injury in vivo. Related KLF family members suppressed or enhanced axon growth to differing extents, and several growth-suppressive KLFs were up-regulated postnatally, whereas growth-enhancing KLFs were down-regulated. Thus, coordinated activities of different KLFs regulate the regenerative capacity of CNS neurons.


Molecular and Cellular Neuroscience | 1999

Distinct neurite outgrowth signaling pathways converge on ERK activation

Jeanette C. Perron; John L. Bixby

Several distinct classes of proteins positively regulate axonal growth; some of these are known to activate the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling cascade, at least in nonneuronal cells. We have found that N-cadherin, as well as laminin (LN) and basic fibroblast growth factor (bFGF), can activate ERK in embryonic chick retinal neurons. Additionally, adhesion of retinal neurons to LN or N-cadherin substrates induced a redistribution of ERK from the cytoplasm toward the plasma membrane. Neurite outgrowth induced by bFGF, LN, or N-cadherin was strongly inhibited by treatment with inhibitors of ERK kinase activation, but not by an inhibitor of p38 MAPK. We conclude (1) that N-cadherin and LN can activate ERK in retinal neurons and (2) that activation of ERK is required for full neurite outgrowth induced by these proteins. Our results suggest that ERK activation is one point of convergence for signaling pathways generated by a variety of axon growth inducers.


Neuron | 1989

Protein kinase C is involved in laminin stimulation of neurite outgrowth

John L. Bixby

We are investigating the intracellular events involved in the induction of neurite outgrowth. The phorbol ester TPA, an activator of protein kinase C, potentiates neurite outgrowth from ciliary ganglion neurons cultured on suboptimal laminin concentrations, but not on optimal laminin concentrations. TPA also stimulates growth on fibronectin and collagen similar to that observed on laminin under control conditions. Manipulations that elevate intracellular cAMP levels (expected to activate A kinase) reduce neurite outgrowth on laminin. The protein kinase C inhibitors H7 and sphingosine inhibit neurite outgrowth on laminin in a reversible and dose-dependent manner. H7 does not inhibit the process outgrowth induced by concanavalin A in the same neurons. The results suggest that activation of protein kinase C is an important step in the neurite outgrowth caused by laminin binding to its receptor(s).


Proceedings of the National Academy of Sciences of the United States of America | 2012

Krüppel-like Factor 7 engineered for transcriptional activation promotes axon regeneration in the adult corticospinal tract

Murray G. Blackmore; Zimei Wang; Jessica K. Lerch; Dario Motti; Yi Ping Zhang; Christopher B. Shields; Jae K. Lee; Jeffrey L. Goldberg; Vance Lemmon; John L. Bixby

Axon regeneration in the central nervous system normally fails, in part because of a developmental decline in the intrinsic ability of CNS projection neurons to extend axons. Members of the KLF family of transcription factors regulate regenerative potential in developing CNS neurons. Expression of one family member, KLF7, is down-regulated developmentally, and overexpression of KLF7 in cortical neurons in vitro promotes axonal growth. To circumvent difficulties in achieving high neuronal expression of exogenous KLF7, we created a chimera with the VP16 transactivation domain, which displayed enhanced neuronal expression compared with the native protein while maintaining transcriptional activation and growth promotion in vitro. Overexpression of VP16-KLF7 overcame the developmental loss of regenerative ability in cortical slice cultures. Adult corticospinal tract (CST) neurons failed to up-regulate KLF7 in response to axon injury, and overexpression of VP16-KLF7 in vivo promoted both sprouting and regenerative axon growth in the CST of adult mice. These findings identify a unique means of promoting CST axon regeneration in vivo by reengineering a developmentally down-regulated, growth-promoting transcription factor.


Neuron | 1995

Agrin is a differentiation-inducing "stop signal" for motoneurons in vitro

Jason A. Campagna; Markus A. Rüegg; John L. Bixby

Proteins of the synaptic basal lamina are important in directing the differentiation of motor nerve terminals. One synaptic basal lamina protein, agrin, which influences postsynaptic muscle differentiation, has been suggested to influence nerve terminals as well. To test this hypothesis, we cocultured chick ciliary ganglion neurons with agrin-expressing CHO cells. Ciliary ganglion neurons, but not sensory neurons, adhered five times as well to agrin-expressing cells as to untransfected cells. Further, ciliary ganglion neurites were growth inhibited upon contact with agrin-expressing cells. Finally, the synaptic vesicle protein synaptotagmin became concentrated at contacts between ciliary ganglion neurites and agrin-expressing cells. These activities were shared by neuronal and muscle-derived agrin isoforms, consistent with the hypothesis that muscle agrin may influence the presynaptic axon. Our results suggest that agrin influences the growth and differentiation of motoneurons in vivo.


Science | 2015

Systemic administration of epothilone B promotes axon regeneration after spinal cord injury

Jörg Ruschel; Farida Hellal; Kevin C. Flynn; Sebastian Dupraz; David A. Elliott; Andrea Tedeschi; Margaret L. Bates; Gary Brook; Kristina Dobrindt; Michael Peitz; Oliver Brüstle; Michael D. Norenberg; Armin Blesch; Norbert Weidner; Mary Bartlett Bunge; John L. Bixby; Frank Bradke

Progress toward fixing a broken back? Axon regeneration after a spinal cord injury requires interference with neuronal mechanisms to promote axon extension and early suppression of scar formation. Microtubule stabilization could provide, in principle, a basis for such intervention. Ruschel et al. used animal models of spinal cord injury, time-lapse imaging in vivo, primary neuronal cultures, and behavioral studies to tackle this challenge (see the Perspective by Tran and Silver). They showed that epothilone B, a U.S. Food and Drug Administration–approved microtubule-stabilizing drug that can cross the blood-brain barrier, does promote functional axon regeneration, even after injury. Science, this issue p. 347; see also p. 285 Stabilizing microtubules after a spinal cord injury reduces the migratory activity of scar-forming meningeal fibroblasts. [Also see Perspective by Tran and Silver] After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier–permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.


Glia | 2008

A new in vitro model of the glial scar inhibits axon growth.

Ina B. Wanner; Andres Deik; Miguel Torres; Andrew Rosendahl; Vance Lemmon; John L. Bixby

Astrocytes respond to central nervous system (CNS) injury with reactive astrogliosis and participate in the formation of the glial scar, an inhibitory barrier for axonal regeneration. Little is known about the injury‐induced mechanisms underlying astrocyte reactivity and subsequent development of an axon‐inhibitory scar. We combined two key aspects of CNS injury, mechanical trauma and co‐culture with meningeal cells, to produce an in vitro model of the scar from cultures of highly differentiated astrocytes. Our model displayed widespread morphological signs of astrocyte reactivity, increases in expression of glial fibrillary acidic protein (GFAP), and accumulation of GFAP in astrocytic processes. Expression levels of scar‐associated markers, phosphacan, neurocan, and tenascins, were also increased. Importantly, neurite growth from various CNS neuronal populations was significantly reduced when neurons were seeded on the scar‐like cultures, compared with growth on cultures of mature astrocytes. Quantification of neurite growth parameters on the scar model demonstrated significant reductions in neuronal adhesion and neurite lengths. Interestingly, neurite outgrowth of postnatal neurons was reduced to a greater extent than that of embryonic neurons, and outgrowth inhibition varied among neuronal populations. Scar‐like reactive sites and neurite‐inhibitory patches were found throughout these cultures, creating a patchwork of growth‐inhibitory areas mimicking a CNS injury site. Thus, our model showed relevant aspects of scar formation and produced widespread inhibition of axonal regeneration; it should be useful both for examining mechanisms underlying scar formation and to assess various treatments for their potential to improve regeneration after CNS injury.


Molecular and Cellular Neuroscience | 2010

High Content Screening of Cortical Neurons Identifies Novel Regulators of Axon Growth

Murray Blackmore; Darcie L. Moore; Robin P. Smith; Jeffrey L. Goldberg; John L. Bixby; Vance Lemmon

Neurons in the central nervous system lose their intrinsic capacity for axon regeneration as they mature, and it is widely hypothesized that changes in gene expression are responsible. Testing this hypothesis and identifying the relevant genes has been challenging because hundreds to thousands of genes are developmentally regulated in CNS neurons, but only a small subset are likely relevant to axon growth. Here we used automated high content analysis (HCA) methods to functionally test 743 plasmids encoding developmentally regulated genes in neurite outgrowth assays using postnatal cortical neurons. We identified both growth inhibitors (Ephexin, Aldolase A, Solute Carrier 2A3, and Chimerin), and growth enhancers (Doublecortin, Doublecortin-like, Kruppel-like Factor 6, and CaM-Kinase II gamma), some of which regulate established growth mechanisms like microtubule dynamics and small GTPase signaling. Interestingly, with only one exception the growth-suppressing genes were developmentally upregulated, and the growth-enhancing genes downregulated. These data provide important support for the hypothesis that developmental changes in gene expression control neurite outgrowth, and identify potential new gene targets to promote neurite outgrowth.


The Journal of Neuroscience | 2005

Receptor Tyrosine Phosphatases Guide Vertebrate Motor Axons during Development

Laurie Stepanek; Andrew W. Stoker; Esther T. Stoeckli; John L. Bixby

Receptor-type protein tyrosine phosphatases (RPTPs) are required for appropriate growth of axons during nervous system development in Drosophila. In the vertebrate, type IIa RPTPs [protein tyrosine phosphatase (PTP)-δ, PTP-σ, and LAR (leukocyte common-antigen-related)] and the type III RPTP, PTP receptor type O (PTPRO), have been implicated in the regulation of axon growth, but their roles in developmental axon guidance are unclear. PTPRO, PTP-δ, and PTP-σ are each expressed in chick motor neurons during the period of axonogenesis. To examine potential roles of RPTPs in axon growth and guidance in vivo, we used double-stranded RNA (dsRNA) interference combined with in ovo electroporation to knock down RPTP expression levels in the embryonic chick lumbar spinal cord. Although most branches of the developing limb nerves appeared grossly normal, a dorsal nerve identified as the anterior iliotibialis was clearly affected by dsRNA knock-down of RPTPs. In experimental embryos treated with dsRNA targeting PTP-δ, PTP-σ, or PTPRO, this nerve showed abnormal fasciculation, was reduced in size, or was missing entirely; interference with PTPRO produced the most severe phenotypes. Control embryos electroporated with vehicle, or with dsRNA targeting choline acetyltransferase or axonin-1, did not exhibit this phenotype. Surprisingly, embryos electroporated with dsRNA targeting PTP-δ together with PTPRO, or all three RPTPs combined, had less severe phenotypes than embryos treated with PTPRO alone. This result suggests that competition between type IIa and type III RPTPs can regulate motor axon outgrowth, consistent with findings in Drosophila. Our results indicate that RPTPs, and especially PTPRO, are required for axon growth and guidance in the developing vertebrate limb.


Science | 2015

Axonal regeneration. Systemic administration of epothilone B promotes axon regeneration after spinal cord injury.

Jörg Ruschel; Farida Hellal; Kevin C. Flynn; Sebastian Dupraz; David A. Elliott; Andrea Tedeschi; Margaret L. Bates; Gary Brook; Kristina Dobrindt; Michael Peitz; Oliver Brüstle; Norenberg; Armin Blesch; Norbert Weidner; Mary Bartlett Bunge; John L. Bixby; Frank Bradke

Progress toward fixing a broken back? Axon regeneration after a spinal cord injury requires interference with neuronal mechanisms to promote axon extension and early suppression of scar formation. Microtubule stabilization could provide, in principle, a basis for such intervention. Ruschel et al. used animal models of spinal cord injury, time-lapse imaging in vivo, primary neuronal cultures, and behavioral studies to tackle this challenge (see the Perspective by Tran and Silver). They showed that epothilone B, a U.S. Food and Drug Administration–approved microtubule-stabilizing drug that can cross the blood-brain barrier, does promote functional axon regeneration, even after injury. Science, this issue p. 347; see also p. 285 Stabilizing microtubules after a spinal cord injury reduces the migratory activity of scar-forming meningeal fibroblasts. [Also see Perspective by Tran and Silver] After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier–permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.

Collaboration


Dive into the John L. Bixby's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge