SNSDP | Home

 

NERVOUS SYSTEM DEVELOPMENT AND PLASTICITY

 

R. Douglas Fields, PhD, Chief, Section on Nervous System Development and Plasticity

Philip Lee, PhD, Research Fellow

Jonathan Cohen, PhD, Postdoctoral Fellow

Tomoko Ishibashi, PhD, Postdoctoral Fellow

Kelly Dakin, BS, Predoctoral fellow

Brian Weinberg, BS, Technician

Beth Stevens, BS, Biologist

 

 

 

Our research is concerned with understanding the molecular mechanisms by which functional activity in the brain regulates development of the nervous system during late stages of fetal development and early postnatal life. The main objectives of our research program are to understand how the expression of genes controlling the developing structure and function of the nervous system are regulated by patterned neural impulse activity and to determine the functional consequences of neural impulse activity on major developmental processes. Areas of study include cell proliferation, survival, differentiation, neurite outgrowth, synaptogenesis and synapse remodeling, myelination, interactions with glia, and the mechanisms of learning and memory.

Activity-dependent neuron-glia interactions

Stevens, Dakin,a Ishibashi, Fields; in collaboration with Gallo

While the importance of neural impulse activity in regulating development of neurons is widely appreciated, such is not the case for development of glia cells, which are non-neuronal. Our recent work has examined the possible influence of axonal impulse activity on developing glia, which form myelin in the peripheral nervous system (PNS) and central nervous system (CNS). Myelin is the spiral wrapping of membrane around axons that provides electrical insulation essential for rapid impulse conduction. Our findings show that distinct purinergic signaling molecules released by axons in an activity-dependent manner control differentiation of myelinating glia in the PNS and CNS.

In the PNS, our studies on Schwann cells show that extracellular ATP released from axons firing action potentials acts on purinergic receptors on Schwann cells (P2Y receptors), causing an increase in intracellular calcium, activation of transcription factors, and regulation of genes involved in Schwann cell differentiation. The result is that several developmental processes in Schwann cells are regulated by the release of ATP from electrically active axons, processes that include inhibiting Schwann cell proliferation, arresting cellular differentiation at an immature stage, and inhibiting myelination.

In studying CNS myelination, our research reveals that oligodendrocyte progenitor cells (OPCs) can detect impulse activity in premyelinated axons and respond with an increase in intracellular calcium. We identified many of the axon-glial signaling molecules involved in impulse activity and found that adenosine, a breakdown product of ATP, is of primary importance. Using RT-PCR, we detected all four types of adenosine receptor in purified cultures of OPCs and in acutely dissociated OPCs from transgenic mice. Impulse activity inhibited cell proliferation of myelinating glia in both the PNS and CNS, but adenosine derived from electrically active axons stimulated OPC differentiation to a promyelinating stage and increased myelination. These findings open new avenues of research into oligodendrocyte development with potential for new therapeutic approaches to treating demyelinating disease.

Fields RD. Opposite effects of impulse activity on myelination in the PNS and CNS via differential axon-glial signaling molecules. Neurochem Int 2003;45:503-509.

Fields RD. The other half of the brain. Sci Am 2004;290:54-61.

Fields RD, Stevens-Graham B. New insights into neuron-glia communication. Science 2002;298:556-562.

Stevens B, Ishibashi T, Chen J-F, Fields RD. Adenosine: an activity-dependent axonal signal regulating MAP kinase and proliferation in developing Schwann cells. Neuron Glia Biol 2004;1:23-34.

Stevens, B, Porta S, Haak LL, Gallo V, Fields RD. Adenosine: a neuron-glial transmitter promoting myelination in the CNS in response to action potentials. Neuron 2002;36:855-868.

Intracellular signaling in conversion of short-term memory into long-term memory

Dudek,b Cohen, Fields

It is widely appreciated that there are two types of memory: short-term and long-term. It has been well known for decades that gene expression is necessary for converting short-term into long-term memory, but it is not known how signals reach the nucleus to initiate the conversion process. Current theory posits that, in response to firing a synapse in the appropriate manner to generate a short-term memory, a signaling molecule generated from the submembrane region of the synapse travels up the dendritic tree to the cell body. After entering the nucleus, the signaling molecule is presumed to initiate gene transcription by activating the transcription factor CREB. The gene product is synthesized and distributed throughout the dendritic tree, but the protein can recognize synapses that have been temporarily strengthened, rendering the change permanent. The identity of this putative synapse-to-nucleus signaling molecule remains unknown.

We proposed an alternative hypothesis based on our studies of action potential–dependent intracellular signaling and CREB activation in DRG neurons. The hypothesis proposed that, rather than generating a specialized synapse-to-nucleus signaling molecule, somatic action potentials could be responsible for activating gene transcription to convert short-term into long-term memory via the same intracellular signaling pathway that we studied in DRG neurons. We tested the hypothesis by causing neurons to fire somatic action potentials in the absence of all excitatory synaptic input in brain slice preparations. Specifically, we backfired axons from CA1 hippocampal neurons in slice preparations in the presence of glutamate neurotransmitter blockers. Following stimulation, we analyzed the slices by immunocytochemistry for activation of CREB and other signaling enzymes involved in long-term potentiation (LTP) of synapses and for expression of genes closely associated with LTP induction. The results showed that backfiring the axons by antidromic stimulation was sufficient to activate CREB and the signaling enzyme MAPK in CA1 neurons in hippocampal slices in the absence of all excitatory synaptic stimulation. When slices were stimulated antidromically, synaptic stimulation that normally results in a transient strengthening of the synapse (e-LTP) resulted instead in long-lasting strengthening of the synapse (l-LTP). The results support our hypothesis and eliminate the necessity of a synapse-to-nucleus signaling molecule for l-LTP and are consistent with the original theory of Hebb, which states that the firing of a neuron determines whether it forms a stronger synaptic connection with another neuron.

Dudek S, Fields RD. Somatic action potentials are sufficient for late-phase LTP-related cell signaling. Proc Natl Acad Sci USA 2002;99:3962-3967.

Gene expression in hippocampal synaptic plasticity

Cohen, Lee, Fields; in collaboration with Becker

Long-term potentiation (LTP) and long-term depression (LTD) are two widely studied forms of synaptic plasticity that can be recorded electrophysiologically in the hippocampus. These phenomena are believed to represent a cellular basis for memory. We are using custom cDNA microarrays to investigate the signaling pathways, genes, and proteins involved in these forms of synaptic plasticity. Our research has identified sets of transcription factors, structural genes, and signaling pathways that are regulated specifically by activity patterns leading selectively to different types of synaptic plasticity. Our objective is to understand how gene-regulatory networks are controlled by the appropriate patterns of impulses leading to different forms of synaptic plasticity and to identify new molecular mechanisms regulating synaptic strength.

Cohen JE, Fields RD. Extracellular calcium depletion in synaptic transmission. Neuroscientist 2003;9:12-17.

Regulation of gene expression by action potential firing patterns

Lee, Cohen, Fields; in collaboration with Becker, Waxman

Using custom cDNA arrays for gene expression profiling, we have investigated how gene expression is regulated by patterned action potential firing in neuronal cell culture. The pattern of impulse activity is regulated by electrical stimulation through platinum electrodes in specially designed cell culture dishes. After stimulation, mRNA and protein expression are measured by gene arrays, quantitative RT-PCR, Western blot, and immunocytochemistry. Our work tests the hypothesis that gene transcription can be regulated by the pattern of action potential firing. Results thus far reveal signaling pathways and gene-regulatory networks that respond selectively to appropriate temporal patterns of action potential firing in neurons. The findings provide a better understanding of how nervous system development and plasticity may be regulated by information encoded in the temporal pattern of impulse firing in the brain.

Fields RD, Lee PR, Cohen JE. Temporal integration of intracellular calcium signaling networks in regulating gene expression by action potentials. Cell Calcium 2004, in press.

Klein JP, Tendi EA, Black JA, Fields RD, Waxman SG. Patterned electrical activity modulates sodium channel expression in sensory neurons. J Neurosci Res 2003;74:192-198.

Neurofibromatosis

Lee, Tendi,c Cohen; in collaboration with Becker, De Vries

Neurofibromatosis (NF-1) is an autosomal dominant condition leading to nerve sheath tumors that can become malignant. We are using gene expression profiling in combination with quantitative RT-PCR and biochemical methods to investigate the processes involved in the transition from a benign to a malignant tumor. We are relying on three types of custom microarrays of genes involved in cell signaling, immune system function, and nervous system expression to compare mRNA expression in normal human Schwann cells and a Schwann cell line derived originally from an NF-1 patient who died from a highly malignant form of neurofibromatosis. Our studies identify hundreds of genes that are dysregulated in the malignant Schwann cells, including those associated with all aspects of cellular function connected with malignancy, such as, for example, those regulating cell proliferation, motility, growth factor, and immune system responses.

Lee PR, Cohen JE, Tendi EA, Farrer R, De Vries GH, Becker KG, Fields RD. Transcriptional profiling in an MPNST-derived cell line and normal human Schwann cells. Neuron Glia Biology 2004, in press.

Stevens B, Fields RD. Regulation of the cell cycle in normal and pathological glia. Neuroscientist 2002;8:93-97.

Stem cells derived from bone marrow for transplantation

Ishibashi

Stem cells can be obtained from several sources in postnatal animals, but the means of controlling their differentiation into the appropriate cell type is a major problem in their use for therapeutic purposes. In an effort to understand the fundamental biological processes controlling differentiation of stem cells into nervous system cells, we are focusing our research on factors controlling differentiation of nervous system cells. We will apply the results in cell culture in animal studies to promote recovery following nervous system injury.

aKelly Dakin, BS, former Postbaccalaureate Fellow

bSevena Dudek, PhD, former Senior Staff Fellow

cElisabetta Tendi, PhD, former Postdoctoral Fellow

COLLABORATORS

Kevin Becker, PhD, Research Resources Branch, NIA, Baltimore, MD

Jiang-Fan Chen, MD, PhD, Boston University School of Medicine, Boston, MA

George De Vries, PhD, Loyola University, Chicago, IL

Vittorio Gallo, PhD, George Washington University, Washington, DC

Michael Schwarzschild, MD, PhD, Massachusetts General Hospital, Charlestown, MA

Stephen Waxman, MD, PhD, Yale University, New Haven, CT


For further information, contact fieldsd@mail.nih.gov