Notch Activity Permits Retinal Cells to Progress Through Multiple Progenitor States and Acquire a Stem Cell Property

Total Page:16

File Type:pdf, Size:1020Kb

Notch Activity Permits Retinal Cells to Progress Through Multiple Progenitor States and Acquire a Stem Cell Property Notch activity permits retinal cells to progress through multiple progenitor states and acquire a stem cell property Ashutosh P. Jadhav*, Seo-Hee Cho†, and Constance L. Cepko†‡ *Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology and †Department of Genetics and Howard Hughes Medical Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115 Contributed by Constance L. Cepko, October 10, 2006 (sent for review November 9, 2005) Signaling through the Notch pathway regulates multiple aspects of Here, we show that activation of Notch in mitotic retinal pro- development. The vertebrate retina allows an investigation of the genitor cells allows the normal temporal progression of progenitor basis for these various effects, because the major cell types of the cells, leading eventually to cells that express features of glial cells, retina arise from a common progenitor that expresses Notch1. The in keeping with previous results (4). However, a comprehensive Notch pathway was constitutively activated in distinct populations molecular characterization revealed that these cells also express of retinal cells during development. Prolonged Notch activity in progenitor genes not expressed by normal Mu¨ller glia. A functional progenitor cells maintained cells in the progenitor state without assay showed that these cells could form neurospheres, as can stem perturbing temporal identity, promoting early progenitor charac- cells, whereas normal retinal cells were unable to form such spheres. teristics early in development and late progenitor characteristics In contrast, when Notch was activated in postmitotic cells, glial cells later in development. Eventually, constitutive Notch activation led without progenitor or stem cell features were produced. these cells to acquire characteristics of glial and stem cells. In contrast, reactivating the Notch pathway in newly postmitotic Results retinal cells promoted mature glial cell formation in a subset of Constitutive Notch Activity in Embryonic Progenitor Cells Promotes cells. These data suggest that prolonged Notch activity does not Formation of Cells with Glial and Progenitor Features in the Postnatal disrupt the normal progression of progenitor temporal states, and Retina. To determine the role of Notch signaling in embryonic that down-regulating or overcoming Notch activity is required for mouse retinal development, the Notch pathway was constitutively proper formation of both neuronal and glial cell fates. activated by crossing RosaN1-IC (17) mice to mice expressing Cre recombinase under the Chx10 promoter (Chx10-CRE; ref. 18). The development ͉ glia ͉ retina RosaN1-IC mice contain, in the ubiquitously expressed Rosa26 locus, a STOP sequence flanked by loxP sites followed by an NIC (Fig. 6A, he generation of cellular diversity during development requires which is published as supporting information on the PNAS web Tthat progenitor cells dynamically interpret both intrinsic and site). Upon exposure to Cre recombinase, the STOP sequence is extrinsic cues (1, 2). Notch is a transmembrane receptor that removed, and NIC is expressed. In addition, an internal ribosomal transduces an extrinsic cue, that of the binding of its ligand, e.g., entry sequence and the gene for nuclear-localized enhanced GFP Delta or Serrate, to directly activate transcription of as-yet ill- leads to GFP expression. This allows for expression of NIC and ϩ defined target genes (3). Studies in both the vertebrate and inver- nuclear GFP in Cre cells and their descendants. Expression of Cre tebrate nervous systems have established a critical role for Notch in Chx10-CRE mice begins at embryonic day (E)11–E12 in retinal signaling in preserving a pool of undifferentiated progenitor cells progenitor cells and is relatively specific to the retina (18). (3). More recent studies have suggested an additional role for The eventual fate of NIC-expressing progenitor cells was assayed at postnatal day 10 (P10), when development is nearly complete. activated Notch in specifying or promoting a particular cell fate, the ϩ glial cell fate (4–6). Furthermore, Notch activity also has been The retinae from NIC-overexpressing mice (Chx10-CRE/ ; N1-IC ϩ N1-IC shown to induce stem cell features and participate in neuronal Rosa / or Chx10 ) were folded and smaller (Fig. 6C) relative to WT retinae (Fig. 6B). Cryosections revealed that the subtype specification (7–9). These studies raise the question of how N1-IC Notch activity can influence the generation of multiple cell fates, Chx10 retina had a slightly smaller outer nuclear layer, ex- while also acting in a more generic fashion to preserve a progenitor panded inner nuclear layer (INL), and reduced inner plexiform pool. layer, causing the ganglion cell layer to compress against the INL (compare Fig. 1 E and F). RNA in situ hybridization (ISH) was The vertebrate retina is a paradigm for studying the mechanisms N1-IC of cell fate determination (1, 10, 11). All major cell types are known performed on Chx10 vs. WT retinae for Notch1, Delta1 (Dll1), to arise sequentially in a conserved order from multipotent pro- Hes1, and Hey1. At this stage, expression of Notch1 and Hes1 is localized to Mu¨ller glia, Hey1 is localized to progenitors (4, 19), and genitors. Studies in frog, rat, chick, and mouse have shown that N1-IC Notch1 is expressed by proliferating and undifferentiated cells Delta1 ISH signal is no longer detectable (13). In Chx10 mice, Notch1 A B during development (12–14). Among the last cells generated in the itself was up-regulated (Fig. 1 vs. ), as were its canonical targets, Hes1 and Hey1, whereas Dll1 was unchanged (data not retina are Mu¨ller glial cells, which retain expression of Notch1 (4, 12). Constitutive activation of the Notch pathway by overexpression of the intracellular domain of Notch (NIC) in fish, frog, chick and Author contributions: A.P.J. and C.L.C. designed research; A.P.J. and S.-H.C. performed rat retina has been shown to inhibit neurogenesis (12, 13, 15, 16). research; A.P.J. contributed new reagents/analytic tools; A.P.J., S.-H.C., and C.L.C. analyzed Furthermore, NIC-overexpressing cells in the fish and rat acquire data; and A.P.J. and C.L.C. wrote the paper. glial features (4, 16), suggesting that Notch may instruct gliogenesis The authors declare no conflict of interest. at the expense of neuronal production. In contrast, activation of the Freely available online through the PNAS open access option. Notch pathway in the early frog retina did not seem to promote Abbreviations: ISH, in situ hybridization; INL, inner nuclear layer; NIC, intracellular domain gliogenesis but resulted in nondividing cells with neuroepithelial of Notch; Pn, postnatal day n;En, embryonic day n. morphology (12). It is unclear whether these differences were due †To whom correspondence should be addressed. E-mail: [email protected]. to the timing and/or dose of prolonged Notch activity. © 2006 by The National Academy of Sciences of the USA 18998–19003 ͉ PNAS ͉ December 12, 2006 ͉ vol. 103 ͉ no. 50 www.pnas.org͞cgi͞doi͞10.1073͞pnas.0608155103 Downloaded by guest on September 28, 2021 However, progenitor genes, such as fibroblast growth factor 15 (FGF15) and cyclin D1 (20, 22), also had higher signals in the Chx10N1-IC retinae (Table 1). To confirm these profiling results, ISH was performed for FGF15 and cyclin D1. Both of these genes are weakly or not expressed at P10, when much of development is complete (20), but were expressed in the NIC-expressing retinae (Fig. 2 A–D). Furthermore, WT and Chx10N1-IC retinae were immunostained for Pax6 and Chx10. At this age, anti-Pax6 antibody marks amacrine, ganglion, and horizontal cells, anti-Chx10 marks bipolar cells and, very weakly, a minority of Mu¨ller glia. During development, both of these genes are expressed in most, if not all, retinal progenitor cells (18, 23). In the WT retina, Pax6 expression can be detected in the lower half of the INL and ganglion cell layer (Fig. 2E), consistent with expression in amacrine and ganglion cells. Interestingly in the Chx10N1-IC retinae, Pax6 was expressed at a high level throughout the INL and found to be colocalized with the GFP-positive cells (Fig. 2F). Chx10 was also found to be expressed at a higher level and colocalized with the NIC-expressing cells (data not shown). Notch Activity Promotes Early Progenitor but Not Late Progenitor or Glial Features in the Embryonic Retina. It was of interest to determine whether Notch activity was sufficient for precocious expression of late progenitor and/or glial genes in early progenitor cells, which normally do not express them. Gene expression profiling was performed on the E13.5 WT and Chx10N1-IC retinae. RNA was isolated from retinae pooled from two WT or mutant mice, and the experiment was performed as a color-swap duplicate. Many of the genes with lower signals in the Chx10N1-IC retinae are markers of retinal ganglion cells, such as GAP43 and neurofilament light polypeptide (NF-L). Because this is the major neuronal cell type Fig. 1. Directed misexpression of NIC in Chxlo expression cells. Section ISH on present at this age, neurogenesis is most likely inhibited in the WT (A and C) and Chx10N1-IC (B and D) retinae at P10. (A and B) Notch1.(C and NIC-overexpressing cells. Genes with higher signals in the N1-IC D) clusterin. Immunostaining on WT (E and I) and Chx10N1-IC (F–H and J–L) Chx10 retinae included Notch1; known Notch targets, such as retinae at P10. (E and F) Merge of DAPI, nGFP, and ␤ tubulin III (␤ tub III). (I and Nrarp (24); and progenitor genes, such as Chx10, FGF15, and Sox9 J) merge of DAPI, nGFP, and glutamine synthetase (glut syn). (G and K) nGFP. (Table 3; ref. 20). Hey1 appeared to have both higher (Table 7) and (H) ␤ tubulin III.
Recommended publications
  • A Rhodopsin Gene Expressed in Photoreceptor Cell R7 of the Drosophila Eye: Homologies with Other Signal-Transducing Molecules
    The Journal of Neuroscience, May 1987, 7(5): 1550-I 557 A Rhodopsin Gene Expressed in Photoreceptor Cell R7 of the Drosophila Eye: Homologies with Other Signal-Transducing Molecules Charles S. Zuker, Craig Montell, Kevin Jones, Todd Laverty, and Gerald M. Rubin Department of Biochemistry, University of California, Berkeley, California 94720 We have isolated an opsin gene from D. melanogaster that example, Pak, 1979; Hardie, 1983; Rubin, 1985). The com- is expressed in the ultraviolet-sensitive photoreceptor cell pound eye of Drosophila contains 3 distinct classesof photo- R7 of the Drosophila compound eye. This opsin gene con- receptor cells, Rl-6, R7, and R8, distinguishableby their mor- tains no introns and encodes a 383 amino acid polypeptide phological arrangement and the spectral behavior of their that is approximately 35% homologous to the blue absorbing corresponding visual pigments (reviewed by Hardie, 1983). In ninaE and Rh2 opsins, which are expressed in photoreceptor each of the approximately 800 ommatidia that make up the eye cells RI-6 and R8, respectively. Amino acid homologies be- there are 6 outer (Rl-R6) and 2 central (1 R7 and 1 R8) pho- tween these different opsins and other signal-transducing toreceptor cells (Fig. 1). The photopigments found in the Rl- molecules suggest an important role for the conserved do- R6 cells, the R7 cell, and the R8 cell differ in their absorption mains of rhodopsin in the transduction of extracellular sig- spectra (Harris et al., 1976) most likely becausedifferent opsin nals. genesare expressedin these distinct classesof photoreceptor cells. The 6 peripheral cells (RI-6) contain the major visual Phototransduction, the neuronal excitation processtriggered by pigment, a rhodopsin that absorbsmaximally at 480 nm (Ostroy light, provides an ideal model system for the study of sensory et al., 1974).
    [Show full text]
  • Chemoreception
    Senses 5 SENSES live version • discussion • edit lesson • comment • report an error enses are the physiological methods of perception. The senses and their operation, classification, Sand theory are overlapping topics studied by a variety of fields. Sense is a faculty by which outside stimuli are perceived. We experience reality through our senses. A sense is a faculty by which outside stimuli are perceived. Many neurologists disagree about how many senses there actually are due to a broad interpretation of the definition of a sense. Our senses are split into two different groups. Our Exteroceptors detect stimulation from the outsides of our body. For example smell,taste,and equilibrium. The Interoceptors receive stimulation from the inside of our bodies. For instance, blood pressure dropping, changes in the gluclose and Ph levels. Children are generally taught that there are five senses (sight, hearing, touch, smell, taste). However, it is generally agreed that there are at least seven different senses in humans, and a minimum of two more observed in other organisms. Sense can also differ from one person to the next. Take taste for an example, what may taste great to me will taste awful to someone else. This all has to do with how our brains interpret the stimuli that is given. Chemoreception The senses of Gustation (taste) and Olfaction (smell) fall under the category of Chemoreception. Specialized cells act as receptors for certain chemical compounds. As these compounds react with the receptors, an impulse is sent to the brain and is registered as a certain taste or smell. Gustation and Olfaction are chemical senses because the receptors they contain are sensitive to the molecules in the food we eat, along with the air we breath.
    [Show full text]
  • Intrinsically Different Retinal Progenitor Cells Produce Specific Types Of
    PERSPECTIVES These clonal data demonstrated that OPINION RPCs are generally multipotent. However, these data could not determine whether Intrinsically different retinal the variability in clones was due to intrinsic differences among RPCs or extrinsic and/ progenitor cells produce specific or stochastic effects on equivalent RPCs or their progeny. Furthermore, the fates identi- fied within a clone demonstrated an RPC’s types of progeny ‘potential’ but not the ability of an RPC to make a specific cell type at a specific devel- Connie Cepko opmental time or its ‘competence’ (BOX 2). Moreover, although many genes that regu- Abstract | Lineage studies conducted in the retina more than 25 years ago late the development of retinal cell types demonstrated the multipotency of retinal progenitor cells (RPCs). The number have been studied, using the now classical and types of cells produced by individual RPCs, even from a single time point in gain- and loss‑of‑function approaches18,19, development, were found to be highly variable. This raised the question of the precise roles of such regulators in defin- whether this variability was due to intrinsic differences among RPCs or to extrinsic ing an RPC’s competence or potential have not been well elucidated, as most studies and/or stochastic effects on equivalent RPCs or their progeny. Newer lineage have examined the outcome of a perturba- studies that have made use of molecular markers of RPCs, retrovirus-mediated tion on the development of a cell type but lineage analyses of specific RPCs and live imaging have begun to provide answers not the stage and/or cell type in which such a to this question.
    [Show full text]
  • The Uvomorulin-Anchorage Protein a Catenin Is a Vinculin
    Proc. Nail. Acad. Sci. USA Vol. 88, pp. 9156-9160, October 1991 Cell Biology The uvomorulin-anchorage protein a catenin is a vinculin homologue KURT HERRENKNECHT*, MASAYUKI OZAWA*t, CHRISTOPH ECKERSKORN*, FRIEDRICH LOTTSPEICHt, MARTIN LENTER*, AND ROLF KEMLER*§ *Max-Planck-Institut ffir Immunbiologie, FG Molekulare Embryologie, D-7800 Freiburg, Federal Republic of Germany; and tMax-Planck-Institut ffr Biochemie, D-8033 Martinsried, Federal Republic of Germany Communicated by Franqois Jacob, July 18, 1991 (receivedfor review June 25, 1991) ABSTRACT The cytoplasmic region of the Ca2+- domain is well conserved in other cadherins, it is possible that dependent cell-adhesion molecule (CAM) uvomorulin associ- catenins may also complex with other members of this gene ates with distinct cytoplasmic proteins with molecular masses family (13, 14). Here we have produced antibodies against a of 102, 88, and 80 kDa termed a, (3, and ycatenin, respectively. catenin and show that a catenin is indeed associated with This complex formation links uvomorulin to the actin filament cadherins from human, mouse, and Xenopus. We have network, which seems to be of primary importance for its cloned and sequenced¶ the cDNA coding for a catenin and cell-adhesion properties. We show here that antibodies against have established the primary protein structure. Sequence a catenin also immunoprecipitate complexes that contain hu- comparison reveals homology to vinculin, a well-known man N-cadherin, mouse P-cadherin, chicken A-CAM (adhe- adherens-type and focal contact protein. rens junction-specific CAM; also called N-cadherin) or Xeno- pus U-cadherin, demonstrating that a catenin is complexed with other cadherins.
    [Show full text]
  • “Análisis De Los Receptores Tirosina Quinasa ALK, RET Y ROS En Los Adenocarcinomas Nasosinusales”
    Universidad de Oviedo Programa de Doctorado “Biomedicina y Oncología Molecular” “Análisis de los receptores tirosina quinasa ALK, RET y ROS en los adenocarcinomas nasosinusales” TESIS DOCTORAL Esteban Reinaldo Pacheco Coronel 20/02/2017 Universidad de Oviedo Programa de Doctorado “Biomedicina y Oncología Molecular” TESIS DOCTORAL “Análisis de los receptores tirosina quinasa ALK, RET y ROS en los adenocarcinomas nasosinusales” Autor: Directores: Esteban Reinaldo José Luís Llorente Pendás Pacheco Coronel Mario Hermsen Dedicatoria A mi familia y amigos, por estar siempre a mi lado y apoyarme en cada momento. Agradecimientos A José Luis por brindarme la oportunidad de trabajar en un tema ambicioso y muy interesante, por los buenos consejos y el tiempo invertido para que este proyecto salga adelante. A Mario, por su valiosa colaboración en el laboratorio, en el análisis de muestras e interpretación de resultados, sus enseñanzas de las diferentes técnicas aplicadas y manejo en el laboratorio; sus consejos sobre la metodología, resultados y conclusiones del proyecto. A mis compañeros del servicio de Otorrinolaringología del Hospital Central de Asturias por sus enseñanzas y el trabajo en equipo. A los compañeros del Instituto Universitario de Oncología del Principado de Asturias. Por que gracias a su trabajo hemos aprendido y desarrollado técnicas importantes para la elaboración de este proyecto. 1 ANTECEDENTES ............................................................................... 1 1.1 Introducción ...................................................................................
    [Show full text]
  • Notch-Signaling in Retinal Regeneration and Müller Glial Plasticity
    Notch-Signaling in Retinal Regeneration and Müller glial Plasticity DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Kanika Ghai, MS Neuroscience Graduate Studies Program The Ohio State University 2009 Dissertation Committee: Dr. Andy J Fischer, Advisor Dr. Heithem El-Hodiri Dr. Susan Cole Dr. Paul Henion Copyright by Kanika Ghai 2009 ABSTRACT Eye diseases such as blindness, age-related macular degeneration (AMD), diabetic retinopathy and glaucoma are highly prevalent in the developed world, especially in a rapidly aging population. These sight-threatening diseases all involve the progressive loss of cells from the retina, the light-sensing neural tissue that lines the back of the eye. Thus, developing strategies to replace dying retinal cells or prolonging neuronal survival is essential to preserving sight. In this regard, cell-based therapies hold great potential as a treatment for retinal diseases. One strategy is to stimulate cells within the retina to produce new neurons. This dissertation elucidates the properties of the primary support cell in the chicken retina, known as the Müller glia, which have recently been shown to possess stem-cell like properties, with the potential to form new neurons in damaged retinas. However, the mechanisms that govern this stem-cell like ability are less well understood. In order to better understand these properties, we analyze the role of one of the key developmental processes, i.e., the Notch-Signaling Pathway in regulating proliferative, neuroprotective and regenerative properties of Müller glia and bestow them with this plasticity.
    [Show full text]
  • Anatomy and Physiology of the Afferent Visual System
    Handbook of Clinical Neurology, Vol. 102 (3rd series) Neuro-ophthalmology C. Kennard and R.J. Leigh, Editors # 2011 Elsevier B.V. All rights reserved Chapter 1 Anatomy and physiology of the afferent visual system SASHANK PRASAD 1* AND STEVEN L. GALETTA 2 1Division of Neuro-ophthalmology, Department of Neurology, Brigham and Womens Hospital, Harvard Medical School, Boston, MA, USA 2Neuro-ophthalmology Division, Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA INTRODUCTION light without distortion (Maurice, 1970). The tear–air interface and cornea contribute more to the focusing Visual processing poses an enormous computational of light than the lens does; unlike the lens, however, the challenge for the brain, which has evolved highly focusing power of the cornea is fixed. The ciliary mus- organized and efficient neural systems to meet these cles dynamically adjust the shape of the lens in order demands. In primates, approximately 55% of the cortex to focus light optimally from varying distances upon is specialized for visual processing (compared to 3% for the retina (accommodation). The total amount of light auditory processing and 11% for somatosensory pro- reaching the retina is controlled by regulation of the cessing) (Felleman and Van Essen, 1991). Over the past pupil aperture. Ultimately, the visual image becomes several decades there has been an explosion in scientific projected upside-down and backwards on to the retina understanding of these complex pathways and net- (Fishman, 1973). works. Detailed knowledge of the anatomy of the visual The majority of the blood supply to structures of the system, in combination with skilled examination, allows eye arrives via the ophthalmic artery, which is the first precise localization of neuropathological processes.
    [Show full text]
  • Specialized Cilia in Mammalian Sensory Systems
    Cells 2015, 4, 500-519; doi:10.3390/cells4030500 OPEN ACCESS cells ISSN 2073-4409 www.mdpi.com/journal/cells Review Specialized Cilia in Mammalian Sensory Systems Nathalie Falk, Marlene Lösl, Nadja Schröder and Andreas Gießl * Department of Biology, Animal Physiology, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; E-Mails: [email protected] (N.F.); [email protected] (M.L.); [email protected] (A.G.) * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +49-9131-85-28055; Fax: +49-9131-85-28060. Academic Editors: Gang Dong and William Tsang Received: 18 May 2015 / Accepted: 9 September 2015 / Published: 11 September 2015 Abstract: Cilia and flagella are highly conserved and important microtubule-based organelles that project from the surface of eukaryotic cells and act as antennae to sense extracellular signals. Moreover, cilia have emerged as key players in numerous physiological, developmental, and sensory processes such as hearing, olfaction, and photoreception. Genetic defects in ciliary proteins responsible for cilia formation, maintenance, or function underlie a wide array of human diseases like deafness, anosmia, and retinal degeneration in sensory systems. Impairment of more than one sensory organ results in numerous syndromic ciliary disorders like the autosomal recessive genetic diseases Bardet-Biedl and Usher syndrome. Here we describe the structure and distinct functional roles of cilia in sensory organs like the inner ear, the olfactory epithelium, and the retina of the mouse. The spectrum of ciliary function in fundamental cellular processes highlights the importance of elucidating ciliopathy-related proteins in order to find novel potential therapies.
    [Show full text]
  • Diversity of Adult Neural Stem and Progenitor Cells in Physiology and Disease
    cells Review Diversity of Adult Neural Stem and Progenitor Cells in Physiology and Disease Zachary Finkel, Fatima Esteban, Brianna Rodriguez, Tianyue Fu, Xin Ai and Li Cai * Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA; [email protected] (Z.F.); [email protected] (F.E.); [email protected] (B.R.); [email protected] (T.F.); [email protected] (X.A.) * Correspondence: [email protected] Abstract: Adult neural stem and progenitor cells (NSPCs) contribute to learning, memory, main- tenance of homeostasis, energy metabolism and many other essential processes. They are highly heterogeneous populations that require input from a regionally distinct microenvironment including a mix of neurons, oligodendrocytes, astrocytes, ependymal cells, NG2+ glia, vasculature, cere- brospinal fluid (CSF), and others. The diversity of NSPCs is present in all three major parts of the CNS, i.e., the brain, spinal cord, and retina. Intrinsic and extrinsic signals, e.g., neurotrophic and growth factors, master transcription factors, and mechanical properties of the extracellular matrix (ECM), collectively regulate activities and characteristics of NSPCs: quiescence/survival, prolifer- ation, migration, differentiation, and integration. This review discusses the heterogeneous NSPC populations in the normal physiology and highlights their potentials and roles in injured/diseased states for regenerative medicine. Citation: Finkel, Z.; Esteban, F.; Keywords: central nervous system (CNS); ependymal cells; neural stem and progenitor cells (NSPC); Rodriguez, B.; Fu, T.; Ai, X.; Cai, L. NG2+ cells; neurodegenerative diseases; regenerative medicine; retina injury; spinal cord injury Diversity of Adult Neural Stem and (SCI); traumatic brain injury (TBI) Progenitor Cells in Physiology and Disease. Cells 2021, 10, 2045.
    [Show full text]
  • Kif1b Rab7a Lmna
    Title: Charcot-Marie-Tooth Neuropathy Type 2 GeneReview Molecular Genetics: Less Commonly Involved Genes Author: Bird TD Updated: March 2016 KIF1B Gene structure. KIF1B comprises 47 exons and 167.13 kb of DNA. Pathogenic allelic variants. See Table A, Locus Specific and HGMD Normal gene product. Kinesin-like protein KIF1B is involved in axonal transport of synaptic vesicle precursors [Zhao et al 2001]. The kinesin superfamily of proteins is essential for intracellular transport along microtubules. Abnormal gene product. There may be a defect in the transport of synaptic vesicles. RAB7A Gene structure. RAB7A has six exons and 87.9 kb of DNA. Pathogenic allelic variants. See Table A. Normal gene product. Ras-related protein Rab-7a belongs to the RAB family of Ras- related GTPases essential for the regulation of intracellular membrane trafficking. Rab- 7a is involved in transport between late endosomes and lysosomes. RAB-interacting lysosomal protein (RILP) induces the recruitment of dynein-dynactin motors and regulates transport toward the minus-end of microtubules [Verhoeven et al 2003]. Abnormal gene product. Abnormal Rab-7a may cause malfunction of lysosomes and inhibit neurite outgrowth [Spinosa et al 2008, Bucci & Deluca 2012]. LMNA Gene structure. LMNA has 12 exons spread over 24 kb of genomic DNA. Pathogenic allelic variants. The most common pathogenic variant found in individuals with CMT2B1 is p.Arg298Cys, a founder mutation in North Africa [Bouhouche et al 2007, De Sandre-Giovannoli et al 2002]. See also Table A. Table 5. Selected LMNA Variants DNA Nucleotide Protein Amino Acid Class of Variant Allele Reference Sequences Change Change Benign c.1908C>T p.= 1 c.398G>T p.Arg133Leu NM_170707.2 c.892C>T p.Arg298Cys Pathogenic NP_733821.1 c.1411C>T p.Arg471Cys c.1579C>T p.Arg527Cys Note on variant classification: Variants listed in the table have been provided by the author.
    [Show full text]
  • Miiller Cells Are a Preferred Substrate for in Vitro Neurite Extension by Rod Photoreceptor Cells
    The Journal of Neuroscience, October 1991, 1 l(10): 2985-2994 Miiller Cells Are a Preferred Substrate for in vitro Neurite Extension by Rod Photoreceptor Cells lvar J. Kljavin’ and Thomas A. Reh2 ‘Neuroscience Research Group, Faculty of Medicine, Lion’s Sight Center, University of Calgary, Calgary, Alberta, Canada T2N lN4 and ‘Department of Biological Structure, University of Washington, Seattle, Washington 98195 To define the factors important in photoreceptor cell mor- (Silver and Sidman, 1980; Krayanek and Goldberg, 1981). Ad- phogenesis, we have examined the ability of rods to extend ditional studies have begun to characterize the moleculesthat neurites in vitro. Retinas from neonatal rats were dissociated are responsiblefor regulating the growth of ganglion cell axons and plated onto substrate-bound extracellular matrix (ECM) during development, and it appearsthat many of the ECM and components or cell monolayers. When rods, identified with cell adhesion molecules involved in axon outgrowth are con- monoclonal antibodies to opsin, were in contact exclusively centrated on the neuroepithelial cell end feet along these long with purified ECM (e.g., laminin, fibronectin, type I collagen, tracts (Silver and Rutishauser, 1984; Halfter et al., 1988). or Matrigel), neurite outgrowth was extremely limited. By However, during the normal development of the retina, as contrast, rods extended long neurites on Miiller cells. Retinal well as other areas of the CNS, most neurons do not extend or brain astrocytes, endothelial cells, 3T3 fibroblasts, or oth- axons into long pathways, but rather, their axons terminate on er retinal neurons were less supportive of rod process out- neighboring cells. In the retina, for example, the axons of the growth.
    [Show full text]
  • Physiology of the Retina
    PHYSIOLOGY OF THE RETINA András M. Komáromy Michigan State University [email protected] 12th Biannual William Magrane Basic Science Course in Veterinary and Comparative Ophthalmology PHYSIOLOGY OF THE RETINA • INTRODUCTION • PHOTORECEPTORS • OTHER RETINAL NEURONS • NON-NEURONAL RETINAL CELLS • RETINAL BLOOD FLOW Retina ©Webvision Retina Retinal pigment epithelium (RPE) Photoreceptor segments Outer limiting membrane (OLM) Outer nuclear layer (ONL) Outer plexiform layer (OPL) Inner nuclear layer (INL) Inner plexiform layer (IPL) Ganglion cell layer Nerve fiber layer Inner limiting membrane (ILM) ©Webvision Inherited Retinal Degenerations • Retinitis pigmentosa (RP) – Approx. 1 in 3,500 people affected • Age-related macular degeneration (AMD) – 15 Mio people affected in U.S. www.nei.nih.gov Mutations Causing Retinal Disease http://www.sph.uth.tmc.edu/Retnet/ Retina Optical Coherence Tomography (OCT) Histology Monkey (Macaca fascicularis) fovea Ultrahigh-resolution OCT Drexler & Fujimoto 2008 9 Adaptive Optics Roorda & Williams 1999 6 Types of Retinal Neurons • Photoreceptor cells (rods, cones) • Horizontal cells • Bipolar cells • Amacrine cells • Interplexiform cells • Ganglion cells Signal Transmission 1st order SPECIES DIFFERENCES!! Photoreceptors Horizontal cells 2nd order Bipolar cells Amacrine cells 3rd order Retinal ganglion cells Visual Pathway lgn, lateral geniculate nucleus Changes in Membrane Potential Net positive charge out Net positive charge in PHYSIOLOGY OF THE RETINA • INTRODUCTION • PHOTORECEPTORS • OTHER RETINAL NEURONS
    [Show full text]