Bio


Rain Runxia Wen obtained her B.Sc. (Hons) in biology in 2012, and her Ph.D. degree in cell and developmental biology in 2018. She joined the Goldberg Lab in the Department of Ophthalmology at Stanford University in 2019. She is currently developing epigenetic tools to promote neuroregeneration, especially retinal ganglion cell, and optic nerve regeneration.

Stanford Advisors


All Publications


  • Kif5a Regulates Mitochondrial Transport in Developing Retinal Ganglion Cells In Vitro. Investigative ophthalmology & visual science Yokota, S., Shah, S. H., Huie, E. L., Wen, R. R., Luo, Z., Goldberg, J. L. 2023; 64 (3): 4

    Abstract

    Axon transport of organelles and neurotrophic factors is necessary for maintaining cellular function and survival of retinal ganglion cells (RGCs). However, it is not clear how trafficking of mitochondria, essential for RGC growth and maturation, changes during RGC development. The purpose of this study was to understand the dynamics and regulation of mitochondrial transport during RGC maturation using acutely purified RGCs as a model system.Primary RGCs were immunopanned from rats of either sex during three stages of development. MitoTracker dye and live-cell imaging were used to quantify mitochondrial motility. Analysis of single-cell RNA sequencing was used to identify Kinesin family member 5A (Kif5a) as a relevant motor candidate for mitochondrial transport. Kif5a expression was manipulated with either short hairpin RNA (shRNA) or exogenous expression adeno-associated virus viral vectors.Anterograde and retrograde mitochondrial trafficking and motility decreased through RGC development. Similarly, the expression of Kif5a, a motor protein that transports mitochondria, also decreased during development. Kif5a knockdown decreased anterograde mitochondrial transport, while Kif5a expression increased general mitochondrial motility and anterograde mitochondrial transport.Our results suggested that Kif5a directly regulates mitochondrial axonal transport in developing RGCs. Future work exploring the role of Kif5a in vivo in RGCs is indicated.

    View details for DOI 10.1167/iovs.64.3.4

    View details for PubMedID 36862119

  • Directly induced human retinal ganglion cells mimic fetal RGCs and are neuroprotective after transplantation invivo. Stem cell reports Luo, Z., Chang, K., Wu, S., Sun, C., Xia, X., Nahmou, M., Bian, M., Wen, R. R., Zhu, Y., Shah, S., Tanasa, B., Wernig, M., Goldberg, J. L. 2022

    Abstract

    Retinal ganglion cell (RGC) replacement therapy could restore vision in glaucoma and other optic neuropathies. We developed a rapid protocol for directly induced RGC (iRGC) differentiation from human stem cells, leveraging overexpression of NGN2. Neuronal morphology and neurite growth were observed within 1week of induction; characteristic RGC-specific gene expression confirmed identity. Calcium imaging demonstrated gamma-aminobutyric acid (GABA)-induced excitation characteristic of immature RGCs. Single-cell RNA sequencing showed more similarities between iRGCs and early-stage fetal human RGCs than retinal organoid-derived RGCs. Intravitreally transplanted iRGCs survived and migrated into host retinas independent of prior optic nerve trauma, but iRGCs protected host RGCs from neurodegeneration. These data demonstrate rapid iRGC generation invitro into an immature cell with high similarity to human fetal RGCs and capacity for retinal integration after transplantation and neuroprotective function after optic nerve injury. The simplicity of this system may benefit translational studies on human RGCs.

    View details for DOI 10.1016/j.stemcr.2022.10.011

    View details for PubMedID 36368332

  • BDNF and cAMP promote retinal ganglion cell survival and function in a porcine model of traumatic optic neuropathy Heng, K., Li, B., Xia, X., Wen, R., Nies, A., Wu, A. Y., Goldberg, J. L. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2022
  • Examination of retrotransposon expression in optic neuropathies Wen, R., Tanasa, B., Xia, X., Nahmou, M., Heng, K., Singh, A., Goldberg, J. L. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2021
  • Intravitreal delivery of AAV2 transduces porcine retinal ganglion cells Heng, K., Li, B., Singh, A., Wen, R., Wu, A. Y., Goldberg, J. L. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2021
  • Autophagy in Xenopus laevis rod photoreceptors is independently regulated by phototransduction and misfolded RHOP23H AUTOPHAGY Wen, R. H., Stanar, P., Tam, B., Moritz, O. L. 2019; 15 (11): 1970–89

    Abstract

    We previously reported autophagic structures in rod photoreceptors expressing a misfolding RHO (rhodopsin) mutant (RHOP23H), suggesting that autophagy may play a role in degrading the mutant RHO and/or be involved in photoreceptor cell death. To further examine autophagy in normal and diseased rods, we generated transgenic Xenopus laevis tadpoles expressing the dually fluorescent autophagy marker mRFP-eGFP-LC3 in rods, which changes from green to yellow and finally red as autophagic structures develop and mature. Using transgenic lines with constitutive and inducible expression, we determined the time-course of autophagy in rod photoreceptors: autophagosomes last for 6 to 8 hours before fusing with lysosomes, and acidified autolysosomes last for about 28 hours before being degraded. Autophagy was diurnally regulated in normal rods, with more autophagic structures generated during periods of light, and this regulation was non-circadian. We also found that more autophagosomes were produced in rods expressing the misfolding RHOP23H mutant. The RHO chromophore absorbs photons to initiate phototransduction, and is consumed in this process; it also promotes RHO folding. To determine whether increased autophagy in light-exposed normal rods is caused by increased RHO misfolding or phototransduction, we used CRISPR/Cas9 to knock out the RPE65 and GNAT1 genes, which are essential for chromophore biosynthesis and phototransduction respectively. Both knockouts suppressed light-induced autophagy, indicating that although light and misfolded rhodopsin can both induce autophagy in rods, light-induced autophagy is not due to misfolding of RHO, but rather due to phototransduction. Abbreviations: CYCS: cytochrome c; bRHOP23H: bovine RHOP23H; Cas9: CRISPR associated protein 9; dpf: days post-fertilization; eGFP: enhanced green fluorescent protein; GNAT1: guanine nucleotide-binding protein G(t) subunit alpha-1 aka rod alpha-transducin; HSPA1A/hsp70: heat shock protein of 70 kilodaltons; LAMP1: lysosomal-associated membrane protein 1; LC3: microtubule-associated protein 1A/1B light chain 3; mRFP: monomeric red fluorescent protein; RHO: rhodopsin; RP: retinitis pigmentosa; RPE65: retinal pigment epithelium-specific 65 kDa protein: sfGFP: superfolding GFP; sgRNA: single guide RNA; WGA: wheat germ agglutinin; RHOp: the Xenopus laevis RHO.2.L promoter.

    View details for DOI 10.1080/15548627.2019.1596487

    View details for Web of Science ID 000466646300001

    View details for PubMedID 30975014

  • Autophagy Induction by HDAC Inhibitors Is Unlikely to be the Mechanism of Efficacy in Prevention of Retinal Degeneration Caused by P23H Rhodopsin Wen, R. H., Loewen, A. D., Vent-Schmidt, R. J., Moritz, O. L., Rickman, C. B., Grimm, C., Anderson, R. E., Ash, J. D., LaVail, M. M., Hollyfield, J. G. SPRINGER INTERNATIONAL PUBLISHING AG. 2019: 401–5

    Abstract

    We previously found that valproic acid (VPA) and other histone deacetylase inhibitors (HDACis) ameliorate retinal degeneration (RD) caused by P23H rhodopsin in Xenopus laevis larvae and hypothesized that this may be due to enhancement of autophagy. Here we use X. laevis expressing an autophagy marker to assess effects of HDACis on autophagy. We also assess the effects of non-HDACi activators and inducers of autophagy on RD caused by P23H rhodopsin.

    View details for DOI 10.1007/978-3-030-27378-1_66

    View details for Web of Science ID 000514087200067

    View details for PubMedID 31884645

  • Opposing Effects of Valproic Acid Treatment Mediated by Histone Deacetylase Inhibitor Activity in Four Transgenic X-laevis Models of Retinitis Pigmentosa JOURNAL OF NEUROSCIENCE Vent-Schmidt, R. J., Wen, R. H., Zong, Z., Chiu, C. N., Tam, B. M., May, C. G., Moritz, O. L. 2017; 37 (4): 1039–54

    Abstract

    Retinitis pigmentosa (RP) is an inherited retinal degeneration (RD) that leads to blindness for which no treatment is available. RP is frequently caused by mutations in Rhodopsin; in some animal models, RD is exacerbated by light. Valproic acid (VPA) is a proposed treatment for RP and other neurodegenerative disorders, with a phase II trial for RP under way. However, the therapeutic mechanism is unclear, with minimal research supporting its use in RP. We investigated the effects of VPA on Xenopus laevis models of RP expressing human P23H, T17M, T4K, and Q344ter rhodopsins, which are associated with RP in humans. VPA ameliorated RD associated with P23H rhodopsin and promoted clearing of mutant rhodopsin from photoreceptors. The effect was equal to that of dark rearing, with no additive effect observed. Rescue of visual function was confirmed by electroretinography. In contrast, VPA exacerbated RD caused by T17M rhodopsin in light, but had no effect in darkness. Effects in T4K and Q344ter rhodopsin models were also negative. These effects of VPA were paralleled by treatment with three additional histone deacetylase (HDAC) inhibitors, but not other antipsychotics, chemical chaperones, or VPA structural analogues. In WT retinas, VPA treatment increased histone H3 acetylation. In addition, electron microscopy showed increased autophagosomes in rod inner segments with HDAC inhibitor (HDACi) treatment, potentially linking the therapeutic effects in P23H rhodopsin animals and negative effects in other models with autophagy. Our results suggest that the success or failure of VPA treatment is dependent on genotype and that HDACi treatment is contraindicated for some RP cases.SIGNIFICANCE STATEMENT Retinitis pigmentosa (RP) is an inherited, degenerative retinal disease that leads to blindness for which no therapy is available. We determined that valproic acid (VPA), currently undergoing a phase II trial for RP, has both beneficial and detrimental effects in animal models of RP depending on the underlying disease mechanism and that both effects are due to histone deacetylase (HDAC) inhibition possibly linked to autophagy regulation. Off-label use of VPA and other HDAC inhibitors for the treatment of RP should be limited to the research setting until this effect is understood and can be predicted. Our study suggests that, unless genotype is accounted for, clinical trials for RP treatments may give negative results due to multiple disease mechanisms with differential responses to therapeutic interventions.

    View details for DOI 10.1523/JNEUROSCI.1647-16.2017

    View details for Web of Science ID 000393569300024

    View details for PubMedID 28490005

    View details for PubMedCentralID PMC6597025

  • Light Induces Ultrastructural Changes in Rod Outer and Inner Segments, Including Autophagy, in a Transgenic Xenopus laevis P23H Rhodopsin Model of Retinitis Pigmentosa INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE Bogea, T. H., Wen, R. H., Moritz, O. L. 2015; 56 (13): 7947–55

    Abstract

    We previously reported a transgenic Xenopus laevis model of retinitis pigmentosa in which tadpoles express the bovine form of P23H rhodopsin (bP23H) in rod photoreceptors. In this model, retinal degeneration was dependent on light exposure. Here, we investigated ultrastructural changes that occurred in the rod photoreceptors of these retinas when exposed to light.Tadpoles expressing bP23H in rods were transferred from constant darkness to a 12-hour light:12-hour dark (12L:12D) regimen. For comparison, transgenic tadpoles expressing an inducible form of caspase 9 (iCasp9) were reared in a 12L:12D regimen, and retinal degeneration was induced by administration of the drug AP20187. Tadpoles were euthanized at various time points, and eyes were processed for confocal light and transmission electron microscopy.We observed defects in outer and inner segments of rods expressing bP23H that were aggravated by light exposure. Rod outer segments exhibited vesiculations throughout and were rapidly phagocytosed by the retinal pigment epithelium. In rod inner segments, we observed autophagic compartments adjacent to the endoplasmic reticulum and extensive vesiculation at later time points. These defects were not found in rods expressing iCasp9, which completely degenerated within 36 hours after drug administration.Our results indicate that ultrastructural defects in outer and inner segment membranes of bP23H expressing rods differ from those observed in drug-induced apoptosis. We suggest that light-induced retinal degeneration caused by P23H rhodopsin occurs via cell death with autophagy, which may represent an attempt to eliminate the mutant rhodopsin and/or damaged cellular compartments from the secretory pathway.

    View details for DOI 10.1167/iovs.15-16799

    View details for Web of Science ID 000368243800041

    View details for PubMedID 26720441

    View details for PubMedCentralID PMC4684193