Bio


Our lab applies biochemical and engineering principles to the development of protein-based tools for imaging and control of biochemical processes. Topics of investigation include fluorescent proteins structure and biophysics, fluorescent protein-based biosensors, neuronal activity sensors, spatiotemporal analysis of protein translation pathways, chemical control of protein translation, and light-responsive proteins.

Academic Appointments


Honors & Awards


  • Burroughs Wellcome Career Award for Medical Scientists, Burroughs Wellcome Foundation (5/2007 - current)

Professional Education


  • BA, Harvard University, Biochemistry (1994)
  • PhD, Harvard Medical School, Biological & Biomedical Sciences, Lab of Michael E. Greenberg (2002)
  • MD, UCLA, Medicine (2004)
  • Postdoctoral Fellowship, UCSD, Lab of Roger Y. Tsien (2009)

Current Research and Scholarly Interests


Our lab applies biochemical and engineering principles to the development of protein-based tools for molecular imaging and gene therapy. Topics of investigation include fluorescent proteins structure and biophysics, fluorescent protein-based biosensors, spatiotemporal analysis of protein translation pathways, chemical control of protein translation, and light-responsive proteins.

2015-16 Courses


Stanford Advisees


Graduate and Fellowship Programs


All Publications


  • High-fidelity optical reporting of neuronal electrical activity with an ultrafast fluorescent voltage sensor NATURE NEUROSCIENCE St-Pierre, F., Marshall, J. D., Yang, Y., Gong, Y., Schnitzer, M. J., Lin, M. Z. 2014; 17 (6): 884-889

    Abstract

    Accurate optical reporting of electrical activity in genetically defined neuronal populations is a long-standing goal in neuroscience. We developed Accelerated Sensor of Action Potentials 1 (ASAP1), a voltage sensor design in which a circularly permuted green fluorescent protein is inserted in an extracellular loop of a voltage-sensing domain, rendering fluorescence responsive to membrane potential. ASAP1 demonstrated on and off kinetics of ∼2 ms, reliably detected single action potentials and subthreshold potential changes, and tracked trains of action potential waveforms up to 200 Hz in single trials. With a favorable combination of brightness, dynamic range and speed, ASAP1 enables continuous monitoring of membrane potential in neurons at kilohertz frame rates using standard epifluorescence microscopy.

    View details for DOI 10.1038/nn.3709

    View details for Web of Science ID 000336638000025

    View details for PubMedID 24755780

  • Non-invasive intravital imaging of cellular differentiation with a bright red-excitable fluorescent protein NATURE METHODS Chu, J., Haynes, R. D., Corbel, S. Y., Li, P., Gonzalez-Gonzalez, E., Burg, J. S., Ataie, N. J., Lam, A. J., Cranfill, P. J., Baird, M. A., Davidson, M. W., Ng, H., Garcia, K. C., Contag, C. H., Shen, K., Blau, H. M., Lin, M. Z. 2014; 11 (5): 572-578

    Abstract

    A method for non-invasive visualization of genetically labeled cells in animal disease models with micrometer-level resolution would greatly facilitate development of cell-based therapies. Imaging of fluorescent proteins (FPs) using red excitation light in the 'optical window' above 600 nm is one potential method for visualizing implanted cells. However, previous efforts to engineer FPs with peak excitation beyond 600 nm have resulted in undesirable reductions in brightness. Here we report three new red-excitable monomeric FPs obtained by structure-guided mutagenesis of mNeptune. Two of these, mNeptune2 and mNeptune2.5, demonstrate improved maturation and brighter fluorescence than mNeptune, whereas the third, mCardinal, has a red-shifted excitation spectrum without reduction in brightness. We show that mCardinal can be used to non-invasively and longitudinally visualize the differentiation of myoblasts into myocytes in living mice with high anatomical detail.

    View details for DOI 10.1038/NMETH.2888

    View details for Web of Science ID 000335873400026

    View details for PubMedID 24633408

  • Optobiology: optical control of biological processes via protein engineering BIOCHEMICAL SOCIETY TRANSACTIONS Kim, B., Lin, M. Z. 2013; 41: 1183-1188

    Abstract

    Enabling optical control over biological processes is a defining goal of the new field of optogenetics. Control of membrane voltage by natural rhodopsin family ion channels has found widespread acceptance in neuroscience, due to the fact that these natural proteins control membrane voltage without further engineering. In contrast, optical control of intracellular biological processes has been a fragmented effort, with various laboratories engineering light-responsive properties into proteins in different manners. In the present article, we review the various systems that have been developed for controlling protein functions with light based on vertebrate rhodopsins, plant photoregulatory proteins and, most recently, the photoswitchable fluorescent protein Dronpa. By allowing biology to be controlled with spatiotemporal specificity and tunable dynamics, light-controllable proteins will find applications in the understanding of cellular and organismal biology and in synthetic biology.

    View details for DOI 10.1042/BST20130150

    View details for Web of Science ID 000324910200011

    View details for PubMedID 24059506

  • Photoswitchable fluorescent proteins: ten years of colorful chemistry and exciting applications CURRENT OPINION IN CHEMICAL BIOLOGY Zhou, X. X., Lin, M. Z. 2013; 17 (4): 682-690

    Abstract

    Reversibly photoswitchable fluorescent proteins (RSFPs) are fluorescent proteins whose fluorescence, upon excitation at a certain wavelength, can be switched on or off by light in a reversible manner. In the last 10 years, many new RSFPs have been developed and novel applications in cell imaging discovered that rely on their photoswitching properties. This review will describe research on the mechanisms of reversible photoswitching and recent applications using RSFPs. While cis-trans isomerization of the chromophore is believed to be the general mechanism for most RSFPs, structural studies reveal diversity in the details of photoswitching mechanisms, including different effects of protonation, chromophore planarity, and pocket flexibility. Applications of RSFPs include new types of live-cell superresolution imaging, tracking of protein movements and interactions, information storage, and optical control of protein activity.

    View details for DOI 10.1016/j.cbpa.2013.05.031

    View details for Web of Science ID 000323585100022

  • Fluorescent and photo-oxidizing TimeSTAMP tags track protein fates in light and electron microscopy NATURE NEUROSCIENCE Butko, M. T., Yang, J., Geng, Y., Kim, H. J., Jeon, N. L., Shu, X., Mackey, M. R., Ellisman, M. H., Tsien, R. Y., Lin, M. Z. 2012; 15 (12): 1742-?

    Abstract

    Protein synthesis is highly regulated throughout nervous system development, plasticity and regeneration. However, tracking the distributions of specific new protein species has not been possible in living neurons or at the ultrastructural level. Previously we created TimeSTAMP epitope tags, drug-controlled tags for immunohistochemical detection of specific new proteins synthesized at defined times. Here we extend TimeSTAMP to label new protein copies by fluorescence or photo-oxidation. Live microscopy of a fluorescent TimeSTAMP tag reveals that copies of the synaptic protein PSD95 are synthesized in response to local activation of growth factor and neurotransmitter receptors, and preferentially localize to stimulated synapses in rat neurons. Electron microscopy of a photo-oxidizing TimeSTAMP tag reveals new PSD95 at developing dendritic structures of immature neurons and at synapses in differentiated neurons. These results demonstrate the versatility of the TimeSTAMP approach for visualizing newly synthesized proteins in neurons.

    View details for DOI 10.1038/nn.3246

    View details for Web of Science ID 000311706700022

    View details for PubMedID 23103964

  • New Alternately Colored FRET Sensors for Simultaneous Monitoring of Zn2+ in Multiple Cellular Locations PLOS ONE Miranda, J. G., Weaver, A. L., Qin, Y., Park, J. G., Stoddard, C. I., Lin, M. Z., Palmer, A. E. 2012; 7 (11)

    Abstract

    Genetically encoded sensors based on fluorescence resonance energy transfer (FRET) are powerful tools for reporting on ions, molecules and biochemical reactions in living cells. Here we describe the development of new sensors for Zn²?based on alternate FRET-pairs that do not involve the traditional CFP and YFP. Zn²? is an essential micronutrient and plays fundamental roles in cell biology. Consequently there is a pressing need for robust sensors to monitor Zn²? levels and dynamics in cells with high spatial and temporal resolution. Here we develop a suite of sensors using alternate FRET pairs, including tSapphire/TagRFP, tSapphire/mKO, Clover/mRuby2, mOrange2/mCherry, and mOrange2/mKATE. These sensors were targeted to both the nucleus and cytosol and characterized and validated in living cells. Sensors based on the new FRET pair Clover/mRuby2 displayed a higher dynamic range and better signal-to-noise ratio than the remaining sensors tested and were optimal for monitoring changes in cytosolic and nuclear Zn²?. Using a green-red sensor targeted to the nucleus and cyan-yellow sensor targeted to either the ER, Golgi, or mitochondria, we were able to monitor Zn²? uptake simultaneously in two compartments, revealing that nuclear Zn²? rises quickly, whereas the ER, Golgi, and mitochondria all sequester Zn²? more slowly and with a delay of 600-700 sec. Lastly, these studies provide the first glimpse of nuclear Zn²? and reveal that nuclear Zn²? is buffered at a higher level than cytosolic Zn²?.

    View details for DOI 10.1371/journal.pone.0049371

    View details for Web of Science ID 000311885300033

    View details for PubMedID 23173058

  • Optical Control of Protein Activity by Fluorescent Protein Domains SCIENCE Zhou, X. X., Chung, H. K., Lam, A. J., Lin, M. Z. 2012; 338 (6108): 810-814

    Abstract

    Fluorescent proteins (FPs) are widely used as optical sensors, whereas other light-absorbing domains have been used for optical control of protein localization or activity. Here, we describe light-dependent dissociation and association in a mutant of the photochromic FP Dronpa, and we used it to control protein activities with light. We created a fluorescent light-inducible protein design in which Dronpa domains are fused to both termini of an enzyme domain. In the dark, the Dronpa domains associate and cage the protein, but light induces Dronpa dissociation and activates the protein. This method enabled optical control over guanine nucleotide exchange factor and protease domains without extensive screening. Our findings extend the applications of FPs from exclusively sensing functions to also encompass optogenetic control.

    View details for DOI 10.1126/science.1226854

    View details for Web of Science ID 000310839500048

    View details for PubMedID 23139335

  • Improving FRET dynamic range with bright green and red fluorescent proteins NATURE METHODS Lam, A. J., St-Pierre, F., Gong, Y., Marshall, J. D., Cranfill, P. J., Baird, M. A., McKeown, M. R., Wiedenmann, J., Davidson, M. W., Schnitzer, M. J., Tsien, R. Y., Lin, M. Z. 2012; 9 (10): 1005-?

    Abstract

    A variety of genetically encoded reporters use changes in fluorescence (or Förster) resonance energy transfer (FRET) to report on biochemical processes in living cells. The standard genetically encoded FRET pair consists of CFPs and YFPs, but many CFP-YFP reporters suffer from low FRET dynamic range, phototoxicity from the CFP excitation light and complex photokinetic events such as reversible photobleaching and photoconversion. We engineered two fluorescent proteins, Clover and mRuby2, which are the brightest green and red fluorescent proteins to date and have the highest Förster radius of any ratiometric FRET pair yet described. Replacement of CFP and YFP with these two proteins in reporters of kinase activity, small GTPase activity and transmembrane voltage significantly improves photostability, FRET dynamic range and emission ratio changes. These improvements enhance detection of transient biochemical events such as neuronal action-potential firing and RhoA activation in growth cones.

    View details for DOI 10.1038/NMETH.2171

    View details for Web of Science ID 000309519300023

    View details for PubMedID 22961245

  • Beyond the rainbow: new fluorescent proteins brighten the infrared scene NATURE METHODS Lin, M. Z. 2011; 8 (9): 726-728

    Abstract

    Two fluorescent proteins that emit in the far-red and infrared range for imaging applications in cells and in vivo are described.

    View details for Web of Science ID 000294439100007

    View details for PubMedID 21878918

  • Toward the Second Generation of Optogenetic Tools JOURNAL OF NEUROSCIENCE Knoepfel, T., Lin, M. Z., Levskaya, A., Tian, L., Lin, J. Y., Boyden, E. S. 2010; 30 (45): 14998-15004

    Abstract

    This mini-symposium aims to provide an integrated perspective on recent developments in optogenetics. Research in this emerging field combines optical methods with targeted expression of genetically encoded, protein-based probes to achieve experimental manipulation and measurement of neural systems with superior temporal and spatial resolution. The essential components of the optogenetic toolbox consist of two kinds of molecular devices: actuators and reporters, which respectively enable light-mediated control or monitoring of molecular processes. The first generation of genetically encoded calcium reporters, fluorescent proteins, and neural activators has already had a great impact on neuroscience. Now, a second generation of voltage reporters, neural silencers, and functionally extended fluorescent proteins hold great promise for continuing this revolution. In this review, we will evaluate and highlight the limitations of presently available optogenic tools and discuss where these technologies and their applications are headed in the future.

    View details for DOI 10.1523/JNEUROSCI.4190-10.2010

    View details for Web of Science ID 000284096300014

    View details for PubMedID 21068304

  • TimeSTAMP tagging of newly synthesized proteins. Current protocols in protein science / editorial board, John E. Coligan ... [et al.] Lin, M. Z., Tsien, R. Y. 2010; Chapter 26: Unit 26 5-?

    Abstract

    The ability to quantify or visualize newly synthesized proteins has important uses in cell biology. For example, a researcher may wish to quantify basal or inducible rates of translation of a specific gene of interest, or detect subcellular locations of newly synthesized copies of a protein in order to study the role of new protein synthesis in the growth of specialized cellular structures. In this unit, the TimeSTAMP method for labeling of newly synthesized copies of a protein of interest is described. In the TimeSTAMP method, the experimenter expresses a protein of interest as a fusion with a cis-acting protease and an epitope tag, both of which are removed by default protease activity. Addition of a specific protease inhibitor then allows preservation of the tag on subsequently synthesized proteins. Finally, the tag is detected by immunological methods.

    View details for DOI 10.1002/0471140864.ps2605s59

    View details for PubMedID 20155731

  • Autofluorescent Proteins with Excitation in the Optical Window for Intravital Imaging in Mammals CHEMISTRY & BIOLOGY Lin, M. Z., McKeown, M. R., Ng, H., Aguilera, T. A., Shaner, N. C., Campbell, R. E., Adams, S. R., Gross, L. A., Ma, W., Alber, T., Tsien, R. Y. 2009; 16 (11): 1169-1179

    Abstract

    Fluorescent proteins have become valuable tools for biomedical research as protein tags, reporters of gene expression, biosensor components, and cell lineage tracers. However, applications of fluorescent proteins for deep tissue imaging in whole mammals have been constrained by the opacity of tissues to excitation light below 600 nm, because of absorbance by hemoglobin. Fluorescent proteins that excite efficiently in the "optical window" above 600 nm are therefore highly desirable. We report here the evolution of far-red fluorescent proteins with peak excitation at 600 nm or above. The brightest one of these, Neptune, performs well in imaging deep tissues in living mice. The crystal structure of Neptune reveals a novel mechanism for red-shifting involving the acquisition of a new hydrogen bond with the acylimine region of the chromophore.

    View details for DOI 10.1016/j.chembiol.2009.10.009

    View details for Web of Science ID 000273207900010

    View details for PubMedID 19942140

  • Mammalian Expression of Infrared Fluorescent Proteins Engineered from a Bacterial Phytochrome SCIENCE Shu, X., Royant, A., Lin, M. Z., Aguilera, T. A., Lev-Ram, V., Steinbach, P. A., Tsien, R. Y. 2009; 324 (5928): 804-807

    Abstract

    Visibly fluorescent proteins (FPs) from jellyfish and corals have revolutionized many areas of molecular and cell biology, but the use of FPs in intact animals, such as mice, has been handicapped by poor penetration of excitation light. We now show that a bacteriophytochrome from Deinococcus radiodurans, incorporating biliverdin as the chromophore, can be engineered into monomeric, infrared-fluorescent proteins (IFPs), with excitation and emission maxima of 684 and 708 nm, respectively; extinction coefficient >90,000 M(-1) cm(-1); and quantum yield of 0.07. IFPs express well in mammalian cells and mice and spontaneously incorporate biliverdin, which is ubiquitous as the initial intermediate in heme catabolism but has negligible fluorescence by itself. Because their wavelengths penetrate tissue well, IFPs are suitable for whole-body imaging. The IFPs developed here provide a scaffold for further engineering.

    View details for DOI 10.1126/science.1168683

    View details for Web of Science ID 000265832400052

    View details for PubMedID 19423828

  • Characterization of Engineered Channel rhodopsin Variants with Improved Properties and Kinetics BIOPHYSICAL JOURNAL Lin, J. Y., Lin, M. Z., Steinbach, P., Tsien, R. Y. 2009; 96 (5): 1803-1814

    Abstract

    Channelrhodopsin 2 (ChR2), a light-activated nonselective cationic channel from Chlamydomonas reinhardtii, has become a useful tool to excite neurons into which it is transfected. The other ChR from Chlamydomonas, ChR1, has attracted less attention because of its proton-selective permeability. By making chimeras of the transmembrane domains of ChR1 and ChR2, combined with site-directed mutagenesis, we developed a ChR variant, named ChEF, that exhibits significantly less inactivation during persistent light stimulation. ChEF undergoes only 33% inactivation, compared with 77% for ChR2. Point mutation of Ile(170) of ChEF to Val (yielding "ChIEF") accelerates the rate of channel closure while retaining reduced inactivation, leading to more consistent responses when stimulated above 25 Hz in both HEK293 cells and cultured hippocampal neurons. In addition, these variants have altered spectral responses, light sensitivity, and channel selectivity. ChEF and ChIEF allow more precise temporal control of depolarization, and can induce action potential trains that more closely resemble natural spiking patterns.

    View details for DOI 10.1016/j.bpj.2008.11.034

    View details for Web of Science ID 000266376500012

    View details for PubMedID 19254539

  • A drug-controllable tag for visualizing newly synthesized proteins in cells and whole animals PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lin, M. Z., Glenn, J. S., Tsien, R. Y. 2008; 105 (22): 7744-7749

    Abstract

    Research on basic cellular processes involving local production or delivery of proteins, such as activity-dependent synaptic modification in neurons, would benefit greatly from a robust, nontoxic method to visualize selectively newly synthesized copies of proteins of interest within cells, tissues, or animals. We report a technique for covalent labeling of newly synthesized proteins of interest based on drug-dependent preservation of epitope tags. Epitope tags are removed from proteins of interest immediately after translation by the activity of a sequence-specific protease until the time a protease inhibitor is added, after which newly synthesized protein copies retain their tags. This method, which we call TimeSTAMP for time-specific tagging for the age measurement of proteins, allows sensitive and nonperturbative visualization and quantification of newly synthesized proteins of interest with exceptionally tight temporal control. We demonstrate applications of TimeSTAMP in retrospectively identifying growing synapses in cultured neurons and in visualizing the distribution of recently synthesized proteins in intact fly brains.

    View details for DOI 10.1073/pnas.0803060105

    View details for Web of Science ID 000256648600023

    View details for PubMedID 18511556

  • Selective labeling of proteins with chemical probes in living cells PHYSIOLOGY Lin, M. Z., Wang, L. 2008; 23 (3): 131-141

    Abstract

    Selective labeling of proteins with small molecules introduces novel chemical and physical properties into proteins, enabling the target protein to be investigated or manipulated with various techniques. Different methods for labeling proteins in living cells have been developed by using protein domains, small peptides, or single amino acids. Their application in cells and in vivo has yielded novel insights into diverse biological processes.

    View details for Web of Science ID 000256685400002

    View details for PubMedID 18556466

  • Improving the photostability of bright monomeric orange and red fluorescent proteins NATURE METHODS Shaner, N. C., Lin, M. Z., McKeown, M. R., Steinbach, P. A., Hazelwood, K. L., Davidson, M. W., Tsien, R. Y. 2008; 5 (6): 545-551

    Abstract

    All organic fluorophores undergo irreversible photobleaching during prolonged illumination. Although fluorescent proteins typically bleach at a substantially slower rate than many small-molecule dyes, in many cases the lack of sufficient photostability remains an important limiting factor for experiments requiring large numbers of images of single cells. Screening methods focusing solely on brightness or wavelength are highly effective in optimizing both properties, but the absence of selective pressure for photostability in such screens leads to unpredictable photobleaching behavior in the resulting fluorescent proteins. Here we describe an assay for screening libraries of fluorescent proteins for enhanced photostability. With this assay, we developed highly photostable variants of mOrange (a wavelength-shifted monomeric derivative of DsRed from Discosoma sp.) and TagRFP (a monomeric derivative of eqFP578 from Entacmaea quadricolor) that maintain most of the beneficial qualities of the original proteins and perform as reliably as Aequorea victoria GFP derivatives in fusion constructs.

    View details for Web of Science ID 000256308200019

    View details for PubMedID 18454154

  • Eph-dependent tyrosine phosphorylation of ephexin1 modulates growth cone collapse NEURON Sahin, M., Greer, P. L., Lin, M. Z., Poucher, H., Eberhart, J., Schmidt, S., Wright, T. M., Shamah, S. M., O'Connel, S., Cowan, C. W., Hu, L., Goldberg, J. L., Debant, A., Corfas, G., Krull, C. E., Greenberg, M. E. 2005; 46 (2): 191-204

    Abstract

    Ephs regulate growth cone repulsion, a process controlled by the actin cytoskeleton. The guanine nucleotide exchange factor (GEF) ephexin1 interacts with EphA4 and has been suggested to mediate the effect of EphA on the activity of Rho GTPases, key regulators of the cytoskeleton and axon guidance. Using cultured ephexin1-/- mouse neurons and RNA interference in the chick, we report that ephexin1 is required for normal axon outgrowth and ephrin-dependent axon repulsion. Ephexin1 becomes tyrosine phosphorylated in response to EphA signaling in neurons, and this phosphorylation event is required for growth cone collapse. Tyrosine phosphorylation of ephexin1 enhances ephexin1's GEF activity toward RhoA while not altering its activity toward Rac1 or Cdc42, thus changing the balance of GTPase activities. These findings reveal that ephexin1 plays a role in axon guidance and is regulated by a switch mechanism that is specifically tailored to control Eph-mediated growth cone collapse.

    View details for DOI 10.1016/j.neuron.2005.01.030

    View details for Web of Science ID 000228674800007

    View details for PubMedID 15848799

  • Survival factor-mediated BAD phosphorylation raises the mitochondrial threshold for apoptosis DEVELOPMENTAL CELL Datta, S. R., Ranger, A. M., Lin, M. Z., Sturgill, J. F., Ma, Y. C., Cowan, C. W., Dikkes, P., Korsmeyer, S. J., Greenberg, M. E. 2002; 3 (5): 631-643

    Abstract

    Growth factor suppression of apoptosis correlates with the phosphorylation and inactivation of multiple proapoptotic proteins, including the BCL-2 family member BAD. However, the physiological events required for growth factors to block cell death are not well characterized. To assess the contribution of BAD inactivation to cell survival, we generated mice with point mutations in the BAD gene that abolish BAD phosphorylation at specific sites. We show that BAD phosphorylation protects cells from the deleterious effects of apoptotic stimuli and attenuates death pathway signaling by raising the threshold at which mitochondria release cytochrome c to induce cell death. These findings establish a function for endogenous BAD phosphorylation, and elucidate a mechanism by which survival kinases block apoptosis in vivo.

    View details for Web of Science ID 000179230900007

    View details for PubMedID 12431371

  • Neurotrophins use the Erk5 pathway to mediate a retrograde survival response NATURE NEUROSCIENCE Watson, F. L., Heerssen, H. M., BHATTACHARYYA, A., Klesse, L., Lin, M. Z., Segal, R. A. 2001; 4 (10): 981-988

    Abstract

    Growth factors synthesized and released by target tissues promote survival and differentiation of innervating neurons. This retrograde signal begins when growth factors bind receptors at nerve terminals. Activated receptors are then endocytosed and transported through the axon to the cell body. Here we show that the mitogen-activated protein kinase (MAPK) signaling pathways used by neurotrophins during retrograde signaling differ from those used following direct stimulation of the cell soma. During retrograde signaling, endocytosed neurotrophin receptors (Trks) activate the extracellular signal-related protein kinase 5 (Erk5) pathway, leading to nuclear translocation of Erk5, phosphorylation of CREB, and enhanced neuronal survival. In contrast, Erk1/2, which mediates nuclear responses following direct cell body stimulation, does not transmit a retrograde signal. Thus, the Erk5 pathway has a unique function in retrograde signaling. Differential activation of distinct MAPK pathways may enable an individual growth factor to relay information that specifies the location and the nature of stimulation.

    View details for Web of Science ID 000171325800011

    View details for PubMedID 11544482

  • EphA receptors regulate growth cone dynamics through the novel guanine nucleotide exchange factor ephexin CELL Shamah, S. M., Lin, M. Z., Goldberg, J. L., Estrach, S., Sahin, M., Hu, L., Bazalakova, M., NEVE, R. L., Corfas, G., Debant, A., Greenberg, M. E. 2001; 105 (2): 233-244

    Abstract

    Eph receptors transduce short-range repulsive signals for axon guidance by modulating actin dynamics within growth cones. We report the cloning and characterization of ephexin, a novel Eph receptor-interacting protein that is a member of the Dbl family of guanine nucleotide exchange factors (GEFs) for Rho GTPases. Ephrin-A stimulation of EphA receptors modulates the activity of ephexin leading to RhoA activation, Cdc42 and Rac1 inhibition, and cell morphology changes. In addition, expression of a mutant form of ephexin in primary neurons interferes with ephrin-A-induced growth cone collapse. The association of ephexin with Eph receptors constitutes a molecular link between Eph receptors and the actin cytoskeleton and provides a novel mechanism for achieving highly localized regulation of growth cone motility.

    View details for Web of Science ID 000168384300010

    View details for PubMedID 11336673

  • Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms CELL Sun, Y., Nadal-Vicens, M., Misono, S., Lin, M. Z., Zubiaga, A., Hua, X. X., Fan, G. P., Greenberg, M. E. 2001; 104 (3): 365-376

    Abstract

    The mechanisms by which neural stem cells give rise to neurons, astrocytes, or oligodendrocytes are beginning to be elucidated. However, it is not known how the specification of one cell lineage results in the suppression of alternative fates. We find that in addition to inducing neurogenesis, the bHLH transcription factor neurogenin (Ngn1) inhibits the differentiation of neural stem cells into astrocytes. While Ngn1 promotes neurogenesis by functioning as a transcriptional activator, Ngn1 inhibits astrocyte differentiation by sequestering the CBP-Smad1 transcription complex away from astrocyte differentiation genes, and by inhibiting the activation of STAT transcription factors that are necessary for gliogenesis. Thus, two distinct mechanisms are involved in the activation and suppression of gene expression during cell-fate specification by neurogenin.

    View details for Web of Science ID 000167003100007

    View details for PubMedID 11239394

  • EphB receptors interact with NMDA receptors and regulate excitatory synapse formation CELL Dalva, M. B., Takasu, M. A., Lin, M. Z., Shamah, S. M., Hu, L., Gale, N. W., Greenberg, M. E. 2000; 103 (6): 945-956

    Abstract

    EphB receptor tyrosine kinases are enriched at synapses, suggesting that these receptors play a role in synapse formation or function. We find that EphrinB binding to EphB induces a direct interaction of EphB with NMDA-type glutamate receptors. This interaction occurs at the cell surface and is mediated by the extracellular regions of the two receptors, but does not require the kinase activity of EphB. The kinase activity of EphB may be important for subsequent steps in synapse formation, as perturbation of EphB tyrosine kinase activity affects the number of synaptic specializations that form in cultured neurons. These findings indicate that EphrinB activation of EphB promotes an association of EphB with NMDA receptors that may be critical for synapse development or function.

    View details for Web of Science ID 000165801300013

    View details for PubMedID 11136979

  • Rapid nuclear responses to target-derived neurotrophins require retrograde transport of ligand-receptor complex JOURNAL OF NEUROSCIENCE Watson, F. L., Heerssen, H. M., Moheban, D. B., Lin, M. Z., Sauvageot, C. M., BHATTACHARYYA, A., Pomeroy, S. L., Segal, R. A. 1999; 19 (18): 7889-7900

    Abstract

    Target-derived neurotrophins initiate signals that begin at nerve terminals and cross long distances to reach the cell bodies and regulate gene expression. Neurotrophin receptors, Trks, themselves serve as retrograde signal carriers. However, it is not yet known whether the retrograde propagation of Trk activation reflects movement of Trk receptors from neurites to cell bodies or reflects serial activation of stationary Trk molecules. Here, we show that neurotrophins selectively applied to distal neurites of sensory neurons rapidly induce phosphorylation of the transcription factor cAMP response element-binding protein (CREB) and also cause a slower increase in Fos protein expression. Both nuclear responses require activation of neurotrophin receptors (Trks) at distal nerve endings and retrograde propagation of Trk activation to the nerve cell bodies. Using photobleach and recovery techniques to follow biologically active, green fluorescent protein (GFP)-tagged BDNF receptors (TrkB-GFP) in live cells during retrograde signaling, we show that TrkB-GFP moves rapidly from neurites to the cell bodies. This rapid movement requires ligand binding, Trk kinase activity, and intact axonal microtubules. When they reach the cell bodies, the activated TrkB receptors are in a complex with ligand. Thus, the retrograde propagation of activated TrkB from neurites to cell bodies, although rapid, reflects microtubule-dependent transport of phosphorylated Trk-ligand complexes. Moreover, the relocation of activated Trk receptors from nerve endings to cell bodies is required for nuclear signaling responses. Together, these data support a model of retrograde signaling whereby rapid vesicular transport of ligand-receptor complex from the neurites to the cell bodies mediates the nuclear responses.

    View details for Web of Science ID 000082539900023

    View details for PubMedID 10479691

  • Akt promotes cell survival by phosphorylating and inhibiting a forkhead transcription factor CELL Brunet, A., Bonni, A., Zigmond, M. J., Lin, M. Z., Juo, P., Hu, L. S., ANDERSON, M. J., Arden, K. C., Blenis, J., Greenberg, M. E. 1999; 96 (6): 857-868

    Abstract

    Survival factors can suppress apoptosis in a transcription-independent manner by activating the serine/ threonine kinase Akt, which then phosphorylates and inactivates components of the apoptotic machinery, including BAD and Caspase 9. In this study, we demonstrate that Akt also regulates the activity of FKHRL1, a member of the Forkhead family of transcription factors. In the presence of survival factors, Akt phosphorylates FKHRL1, leading to FKHRL1's association with 14-3-3 proteins and FKHRL1's retention in the cytoplasm. Survival factor withdrawal leads to FKHRL1 dephosphorylation, nuclear translocation, and target gene activation. Within the nucleus, FKHRL1 triggers apoptosis most likely by inducing the expression of genes that are critical for cell death, such as the Fas ligand gene.

    View details for Web of Science ID 000079300100012

    View details for PubMedID 10102273