1
|
Gunewardene N, Lam P, Song J, Nguyen T, Ruiz SM, Wong RCB, Wise AK, Richardson RT. Extent of genetic and epigenetic factor reprogramming via a single viral vector construct in deaf adult mice. Hear Res 2025; 457:109170. [PMID: 39848037 DOI: 10.1016/j.heares.2024.109170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025]
Abstract
In the adult mammalian cochlea, hair cell loss is irreversible and causes deafness. The basic helix-loop transcription factor Atoh1 is essential for normal hair cell development in the embryonic ear. Over-expression of Atoh1 in the adult cochlea by gene therapy can convert supporting cells (cells that underlie hair cells) into a hair cell lineage. However, the regeneration outcomes can be inconsistent. Given that hair cell development is regulated by multiple signalling and transcriptional factors in a temporal and spatial manner, a more complex combinatorial approach targeting additional transcription factors may be required for efficient hair cell regeneration. There is evidence that epigenetic factors are responsible for the lack in regenerative capacity of the deaf adult cochlea. This study aimed to develop a combined gene therapy approach to reprogram both the genome and epigenome of supporting cells to improve the efficiency of hair cell regeneration. Adult Pou4f3-DTR mice were used in which the administration of diphtheria toxin was used to ablate hair cells whilst leaving supporting cells relatively intact. A single adeno-associated viral construct was used to express human Atoh1, Pou4f3 and short hairpin RNA against Kdm1a (regeneration gene therapy) at two weeks following partial or severe hair cell ablation. The average transduction of the inner supporting cells, as measured by the control AAV2.7m8-GFP vector in the deaf cochlea, was only 8 % while transduction in the outer sensory region was <1 %. At 4- and 6-weeks post-treatment the number of Myo+ hair cells in the control and regeneration gene therapy-treated mice were not significantly different. Of note, although both control and regeneration gene therapy treated cochleae contained supporting cells that co-expressed the hair cell marker Myo7a and the supporting cell marker Sox2, the regeneration gene therapy treated cochleae had significantly higher numbers of these cells (p < 0.05). Furthermore, among these treated cochleae, those that had more hair cell loss had a higher number of Myo7a positive supporting cells (R2=0.33, Pearson correlation analysis, p < 0.001). Overall, our results indicate that the adult cochlea possesses limited intrinsic spontaneous regenerative capacity, that can be further enhanced by genetic and epigenetic reprogramming.
Collapse
Affiliation(s)
- Niliksha Gunewardene
- Bionics Institute, East Melbourne, Victoria 3002, Australia; Department of Medical Bionics, The University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Patrick Lam
- Bionics Institute, East Melbourne, Victoria 3002, Australia
| | - Jiwei Song
- Bionics Institute, East Melbourne, Victoria 3002, Australia
| | - Trung Nguyen
- Bionics Institute, East Melbourne, Victoria 3002, Australia
| | - Shannon Mendez Ruiz
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, Victoria, Australia
| | - Raymond C B Wong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, Victoria, Australia
| | - Andrew K Wise
- Bionics Institute, East Melbourne, Victoria 3002, Australia; Department of Medical Bionics, The University of Melbourne, Fitzroy, Victoria 3065, Australia; Department of Surgery (Otolaryngology), University of Melbourne, The Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria 3002, Australia
| | - Rachael T Richardson
- Bionics Institute, East Melbourne, Victoria 3002, Australia; Department of Medical Bionics, The University of Melbourne, Fitzroy, Victoria 3065, Australia; Department of Surgery (Otolaryngology), University of Melbourne, The Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria 3002, Australia.
| |
Collapse
|
2
|
Walters AS, Li Y, Karroum EG, Champion D, Weinstock LB, Bagai K, Afrin LB, Spruyt K. Exploring the role of the endogenous opiate system in the pathogenesis of anemia in an opiate receptor knock-out model of Restless Legs Syndrome. Med Hypotheses 2022; 167:110941. [PMID: 36505961 PMCID: PMC9731170 DOI: 10.1016/j.mehy.2022.110941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Restless Legs Syndrome (RLS) is characterized by bothersome leg discomfort accompanied by an urge to move to obtain relief and symptoms are worse at night and on lying down. There is at least partial and temporary relief with activity. It is also an opioid responsive disorder, often accompanied by iron deficiency with or without anemia, and inflammation may be a precipitating factor in some cases. We created two in-vivo opiate receptor knock out mouse models of RLS - a triple opiate receptor knock-out mouse and a mu opiate receptor knock-out mouse. Both sets of animals were restless during the sleep period as is also true of RLS. Both of our knockout models showed statistically significantly decreased Hemoglobin and Hematocrit indicating anemia and both models showed statistically significant decreases in serum iron suggestive of either iron deficiency anemia or inflammatory anemia. The rest of the hematologic studies were not consistent enough to determine which of these two types of anemia was present in either model. An additional experiment in normal wild type mice showed a statistically significant decrease in serum iron when an opiate receptor blocker was used. To our knowledge this is the first demonstration that deficiency of endogenous opioids might play a role in the production of anemia. Our hypothesis is that an intact endogenous opiate system is necessary for red cell homeostasis. The presence of opioid receptors both on red blood cells and on various immunologically based white blood cells suggest mechanisms by which deficiency in the endogenous opiate system could cause anemia of either the iron deficiency or inflammatory types. The administration of opioid agonists or antagonists to iron deficient cultures of red blood cell precursors is a next step in determining the role of the endogenous opiate system in the maintenance of red cell homeostasis and in the possible prevention of iron deficiency or inflammatory anemia where iron dysregulation is key.
Collapse
Affiliation(s)
- Arthur S. Walters
- Sleep Division, Dept of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Dept of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Elias G. Karroum
- Department of Neurology & Rehabilitation Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC., USA
| | - David Champion
- Sydney Children’s Hospital, Randwick, NSW 2031, Australia
| | - Leonard B. Weinstock
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kanika Bagai
- Sleep Division, Dept of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lawrence B. Afrin
- Hematology/Oncology, AIM Center for Personalized Medicine, Purchase, New York
| | - Karen Spruyt
- Université de Paris, NeuroDiderot Inserm, France
| |
Collapse
|
3
|
Peng H, Ramadurgum P, Woodard DR, Daniel S, Nakahara E, Renwick M, Aredo B, Datta S, Chen B, Ufret-Vincenty R, Hulleman JD. Utility of the DHFR-based destabilizing domain across mouse models of retinal degeneration and aging. iScience 2022; 25:104206. [PMID: 35521529 PMCID: PMC9062244 DOI: 10.1016/j.isci.2022.104206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 02/16/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
The Escherichia coli dihydrofolate reductase (DHFR) destabilizing domain (DD) serves as a promising approach to conditionally regulate protein abundance in a variety of tissues. To test whether this approach could be effectively applied to a wide variety of aged and disease-related ocular mouse models, we evaluated the DHFR DD system in the eyes of aged mice (up to 24 months), a light-induced retinal degeneration (LIRD) model, and two genetic models of retinal degeneration (rd2 and Abca4−/− mice). The DHFR DD was effectively degraded in all model systems, including rd2 mice, which showed significant defects in chymotrypsin proteasomal activity. Moreover, trimethoprim (TMP) administration stabilized the DHFR DD in all mouse models. Thus, the DHFR DD-based approach allows for control of protein abundance in a variety of mouse models, laying the foundation to use this strategy for the conditional control of gene therapies to potentially treat multiple eye diseases. Destabilizing domains (DDs) confer conditional control of ocular protein abundance The DHFR DD is effectively turned over and stabilized in aged mouse’s retina DHFR DDs perform well in environmental and genetic retinal degenerative models
Collapse
|
4
|
Regulation of protein secretion through chemical regulation of endoplasmic reticulum retention signal cleavage. Nat Commun 2022; 13:1323. [PMID: 35260576 PMCID: PMC8904541 DOI: 10.1038/s41467-022-28971-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Secreted proteins, such as hormones or cytokines, are key mediators in multicellular organisms. Response of protein secretion based on transcriptional control is rather slow, as it requires transcription, translation and transport from the endoplasmic reticulum (ER) to the plasma membrane via the conventional protein secretion (CPS) pathway. An alternative regulation to provide faster response would be valuable. Here we present two genetically encoded orthogonal regulatory secretion systems, which rely on the retention of pre-synthesized proteins on the ER membrane (membER, released by a cytosolic protease) or inside the ER lumen (lumER, released by an ER-luminal protease), respectively, and their release by the chemical signal-regulated proteolytic removal of an ER-retention signal, without triggering ER stress due to protein aggregates. Design of orthogonal chemically-regulated split proteases enables the combination of signals into logic functions. Its application was demonstrated on a chemically regulated therapeutic protein secretion and regulated membrane translocation of a chimeric antigen receptor (CAR) targeting cancer antigen. Regulation of the ER escape represents a platform for the design of fast-responsive and tightly-controlled modular and scalable protein secretion system for mammalian cells. Secreted proteins, such as hormones or cytokines, are key mediators in multicellular organisms. Here the authors present two genetically encoded orthogonal regulatory secretion systems that enables inducible protein release and construction of logic gates.
Collapse
|
5
|
Tornabene P, Trapani I, Centrulo M, Marrocco E, Minopoli R, Lupo M, Iodice C, Gesualdo C, Simonelli F, Surace EM, Auricchio A. Inclusion of a degron reduces levelsof undesired inteins after AAV-mediated protein trans-splicing in the retina. Mol Ther Methods Clin Dev 2021; 23:448-459. [PMID: 34786437 PMCID: PMC8571531 DOI: 10.1016/j.omtm.2021.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 12/02/2022]
Abstract
Split intein-mediated protein trans-splicing expands AAV transfer capacity, thus overcoming the limited AAV cargo. However, non-mammalian inteins persist as trans-splicing by-products, and this could raise safety concerns for AAV intein clinical applications. In this study, we tested the ability of several degrons to selectively decrease levels of inteins after protein trans-splicing and found that a version of E. coli dihydrofolate reductase, which we have shortened to better fit into the AAV vector, is the most effective. We show that subretinal administration of AAV intein armed with this short degron is both safe and effective in a mouse model of Stargardt disease (STGD1), which is the most common form of inherited macular degeneration in humans. This supports the use of optimized AAV intein for gene therapy of both STGD1 and other conditions that require transfer of large genes.
Collapse
Affiliation(s)
- Patrizia Tornabene
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples 80131, Italy
| | - Miriam Centrulo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Renato Minopoli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Mariangela Lupo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Carolina Iodice
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Carlo Gesualdo
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania L. Vanvitelli, Naples 80131, Italy
| | - Francesca Simonelli
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania L. Vanvitelli, Naples 80131, Italy
| | - Enrico M. Surace
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples 80131, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
- Medical Genetics, Department of Advanced Biomedicine, Federico II University, Naples 80131, Italy
- Correspondence: Alberto Auricchio, MD, Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy; Medical Genetics, Department of Translational Medicine, Federico II University, Naples 80131, Italy.
| |
Collapse
|
6
|
Sun J, Zhang W, Zhao Y, Liu J, Wang F, Han Y, Jiang M, Li S, Tang D. Conditional control of chimeric antigen receptor T-cell activity through a destabilizing domain switch and its chemical ligand. Cytotherapy 2021; 23:1085-1096. [PMID: 34593327 DOI: 10.1016/j.jcyt.2021.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AIMS Despite the impressive efficacy of chimeric antigen receptor (CAR) T-cell therapy, adverse effects, including cytokine release syndrome and neurotoxicity, impede its therapeutic application, thus making the modulation of CAR T-cell activity a priority. The destabilizing domain mutated from Escherichia coli dihydrofolate reductase (DHFR) is inherently unstable and degraded by proteasomes unless it is stabilized by its chemical ligand trimethoprim (TMP), a Food and Drug Administration-approved drug. Here the authors reveal a strategy to modulate CAR T-cell activity at the protein level by employing DHFR and TMP as a chemical switch system. METHODS First, the system was demonstrated to work in human primary T cells. To introduce the system to CAR T cells, DHFR was genetically fused to the carboxyl terminal of a third-generation CAR molecule targeting CD19 (CD19-CAR), constructing the CD19-CAR-DHFR fusion. RESULTS The CD19-CAR-DHFR molecule level was shown to be modulated by TMP. Importantly, the incorporation of DHFR had no impact on the recognition specificity and normal function of the CAR molecule. Little adverse effect on cell proliferation and apoptosis was detected. It was proved that TMP could regulate cytokine secretion and the in vitro cytotoxicity of CD19-CAR-DHFR T cells. Furthermore, the in vivo anti-tumor efficacy was demonstrated to be controllable through the manipulation of TMP administration. The approach to control CD19-CAR also succeeded in 19-BBZ(71), another CD19-targeting CAR with a different structure. CONCLUSIONS The proposed approach based on DHFR and TMP provides a facile strategy to bring CAR T-cell therapy under conditional user control, and the strategy may have the potential to be transplantable.
Collapse
Affiliation(s)
- Jiao Sun
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Wen Zhang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China.
| | - Yi Zhao
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Jiang Liu
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Fang Wang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Ying Han
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Miao Jiang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Shiwu Li
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Dongqi Tang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
7
|
Izert MA, Klimecka MM, Górna MW. Applications of Bacterial Degrons and Degraders - Toward Targeted Protein Degradation in Bacteria. Front Mol Biosci 2021; 8:669762. [PMID: 34026843 PMCID: PMC8138137 DOI: 10.3389/fmolb.2021.669762] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/15/2021] [Indexed: 12/28/2022] Open
Abstract
A repertoire of proteolysis-targeting signals known as degrons is a necessary component of protein homeostasis in every living cell. In bacteria, degrons can be used in place of chemical genetics approaches to interrogate and control protein function. Here, we provide a comprehensive review of synthetic applications of degrons in targeted proteolysis in bacteria. We describe recent advances ranging from large screens employing tunable degradation systems and orthogonal degrons, to sophisticated tools and sensors for imaging. Based on the success of proteolysis-targeting chimeras as an emerging paradigm in cancer drug discovery, we discuss perspectives on using bacterial degraders for studying protein function and as novel antimicrobials.
Collapse
Affiliation(s)
| | | | - Maria Wiktoria Górna
- Structural Biology Group, Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Ineichen BV, Di Palma S, Laczko E, Liddelow SA, Neumann S, Schwab ME, Mosberger AC. Regional Differences in Penetration of the Protein Stabilizer Trimethoprim (TMP) in the Rat Central Nervous System. Front Mol Neurosci 2020; 13:167. [PMID: 33013318 PMCID: PMC7496896 DOI: 10.3389/fnmol.2020.00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/11/2020] [Indexed: 12/04/2022] Open
Abstract
Regulating gene expression at the protein level is becoming increasingly important for answering basic questions in neurobiology. Several techniques using destabilizing domains (DD) on transgenes, which can be activated or deactivated by specific drugs, have been developed to achieve this goal. A DD from bacterial dihydrofolate reductase bound and stabilized by trimethoprim (TMP) represents such a tool. To control transgenic protein levels in the brain, the DD-regulating drugs need to have sufficient penetration into the central nervous system (CNS). Yet, very limited information is available on TMP pharmacokinetics in the CNS following systemic injection. Here, we performed a pharmacokinetic study on the penetration of TMP into different CNS compartments in the rat. We used mass spectrometry to measure TMP concentrations in serum, cerebrospinal fluid (CSF) and tissue samples of different CNS regions upon intraperitoneal TMP injection. We show that TMP quickly (within 10 min) penetrates from serum to CSF through the blood-CSF barrier. TMP also shows quick penetration into brain tissue but concentrations were an order of magnitude lower compared to serum or CSF. TMP concentration in spinal cord was lower than in any other analyzed CNS area. Nevertheless, effective levels of TMP to stabilize DDs can be reached in the CNS with half-lives around 2 h. These data show that TMP has good and fast penetration properties into the CNS and is therefore a valuable ligand for precisely controlling protein expression in the CNS in rodents.
Collapse
Affiliation(s)
- Benjamin V Ineichen
- Department of Health Sciences and Technology, Brain Research Institute, University of Zurich, ETH Zürich, Zurich, Switzerland
| | - Serena Di Palma
- Functional Genomics Center Zurich, University of Zurich, ETH Zürich, Zurich, Switzerland
| | - Endre Laczko
- Functional Genomics Center Zurich, University of Zurich, ETH Zürich, Zurich, Switzerland
| | - Shane A Liddelow
- Neuroscience Institute, NYU School of Medicine, New York, NY, United States.,Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, United States.,Department of Ophthalmology, NYU School of Medicine, New York, NY, United States
| | - Susanne Neumann
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin E Schwab
- Department of Health Sciences and Technology, Brain Research Institute, University of Zurich, ETH Zürich, Zurich, Switzerland
| | - Alice C Mosberger
- Department of Health Sciences and Technology, Brain Research Institute, University of Zurich, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
9
|
Ramadurgum P, Daniel S, Hulleman JD. Protocol for In Vivo Evaluation and Use of Destabilizing Domains in the Eye, Liver, and Beyond. STAR Protoc 2020; 1. [PMID: 32995756 PMCID: PMC7521670 DOI: 10.1016/j.xpro.2020.100094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Destabilizing domains (DDs) have been used successfully to conditionally control the abundance of proteins of interest (POIs) in a small-molecule-dependent manner in mice, worms (Caenorhabditis elegans), and Drosophila. However, development of such systems must account for delivery of the DD-POIs to the target tissue, accessibility of the target tissue to the small molecule, and quantification of stabilization. Here, we describe the considerations and steps to take in order to effectively implement a DD-POI in mouse ocular and hepatic tissue. For complete details on the use and execution of this protocol, please refer to Datta et al. (2018), Ramadurgum and Hulleman (2020), and Ramadurgum et al. (2020).
Collapse
Affiliation(s)
- Prerana Ramadurgum
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Steffi Daniel
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - John D Hulleman
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.,Technical Contact
| |
Collapse
|
10
|
Carvalho VMDA, Nakahara TS, Souza MADA, Cardozo LM, Trintinalia GZ, Pissinato LG, Venancio JO, Stowers L, Papes F. Representation of Olfactory Information in Organized Active Neural Ensembles in the Hypothalamus. Cell Rep 2020; 32:108061. [PMID: 32846119 DOI: 10.1016/j.celrep.2020.108061] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/20/2020] [Accepted: 07/31/2020] [Indexed: 11/17/2022] Open
Abstract
The internal representation of sensory information via coherent activation of specific pathways in the nervous system is key to appropriate behavioral responses. Little is known about how chemical stimuli that elicit instinctive behaviors lead to organized patterns of activity in the hypothalamus. Here, we study how a wide range of chemosignals form a discernible map of olfactory information in the ventromedial nucleus of the hypothalamus (VMH) and show that different stimuli entail distinct active neural ensembles. Importantly, we demonstrate that this map depends on functional inputs from the vomeronasal organ. We present evidence that the spatial locations of active VMH ensembles are correlated with activation of distinct vomeronasal receptors and that disjunct VMH ensembles exhibit differential projection patterns. Moreover, active ensembles with distinct spatial locations are not necessarily associated with different behavior categories, such as defensive or social, calling for a revision of the currently accepted model of VMH organization.
Collapse
Affiliation(s)
- Vinicius Miessler de Andrade Carvalho
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Department of Cell Biology, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Thiago Seike Nakahara
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Mateus Augusto de Andrade Souza
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Leonardo Minete Cardozo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Guilherme Ziegler Trintinalia
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Leonardo Granato Pissinato
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - José Otávio Venancio
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil
| | - Lisa Stowers
- Department of Cell Biology, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Fabio Papes
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862, Brazil.
| |
Collapse
|
11
|
Simultaneous Control of Endogenous and User-Defined Genetic Pathways Using Unique ecDHFR Pharmacological Chaperones. Cell Chem Biol 2020; 27:622-634.e6. [PMID: 32330442 DOI: 10.1016/j.chembiol.2020.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/04/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022]
Abstract
Destabilizing domains (DDs), such as a mutated form of Escherichia coli dihydrofolate reductase (ecDHFR), confer instability and promote protein degradation. However, when combined with small-molecule stabilizers (e.g., the antibiotic trimethoprim), DDs allow positive regulation of fusion protein abundance. Using a combinatorial screening approach, we identified and validated 17 unique 2,4-diaminopyrimidine/triazine-based ecDHFR DD stabilizers, at least 15 of which were ineffective antibiotics against E. coli and S. aureus. Identified stabilizers functioned in vivo to control an ecDHFR DD-firefly luciferase in the mouse eye and/or the liver. Next, stabilizers were leveraged to perform synergistic dual functions in vitro (HeLa cell death sensitization) and in vivo (repression of ocular inflammation) by stabilizing a user-defined ecDHFR DD while also controlling endogenous signaling pathways. Thus, these newly identified pharmacological chaperones allow for simultaneous control of compound-specific endogenous and user-defined genetic pathways, the combination of which may provide synergistic effects in complex biological scenarios.
Collapse
|
12
|
Peng H, Chau VQ, Phetsang W, Sebastian RM, Stone MRL, Datta S, Renwick M, Tamer YT, Toprak E, Koh AY, Blaskovich MA, Hulleman JD. Non-antibiotic Small-Molecule Regulation of DHFR-Based Destabilizing Domains In Vivo. Mol Ther Methods Clin Dev 2019; 15:27-39. [PMID: 31649953 PMCID: PMC6804886 DOI: 10.1016/j.omtm.2019.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
The E. coli dihydrofolate reductase (DHFR) destabilizing domain (DD), which shows promise as a biologic tool and potential gene therapy approach, can be utilized to achieve spatial and temporal control of protein abundance in vivo simply by administration of its stabilizing ligand, the routinely prescribed antibiotic trimethoprim (TMP). However, chronic TMP use drives development of antibiotic resistance (increasing likelihood of subsequent infections) and disrupts the gut microbiota (linked to autoimmune and neurodegenerative diseases), tempering translational excitement of this approach in model systems and for treating human diseases. Herein, we identified a TMP-based, non-antibiotic small molecule, termed 14a (MCC8529), and tested its ability to control multiple DHFR-based reporters and signaling proteins. We found that 14a is non-toxic and can effectively stabilize DHFR DDs expressed in mammalian cells. Furthermore, 14a crosses the blood-retinal barrier and stabilizes DHFR DDs expressed in the mouse eye with kinetics comparable to that of TMP (≤6 h). Surprisingly, 14a stabilized a DHFR DD in the liver significantly better than TMP did, while having no effect on the mouse gut microbiota. Our results suggest that alternative small-molecule DHFR DD stabilizers (such as 14a) may be ideal substitutes for TMP in instances when conditional, non-antibiotic control of protein abundance is desired in the eye and beyond.
Collapse
Affiliation(s)
- Hui Peng
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Viet Q. Chau
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Wanida Phetsang
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, QLD 4072, Australia
| | - Rebecca M. Sebastian
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - M. Rhia L. Stone
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, QLD 4072, Australia
| | - Shyamtanu Datta
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Marian Renwick
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Yusuf T. Tamer
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Erdal Toprak
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Andrew Y. Koh
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Mark A.T. Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, QLD 4072, Australia
| | - John D. Hulleman
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
13
|
Small Molecule-Based Inducible Gene Therapies for Retinal Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1185:65-69. [PMID: 31884590 DOI: 10.1007/978-3-030-27378-1_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The eye is an excellent target organ for gene therapy. It is physically isolated, easily accessible, immune-privileged, and postmitotic. Furthermore, potential gene therapies introduced into the eye can be evaluated by noninvasive methods such as fundoscopy, electroretinography, and optical coherence tomography. In the last two decades, great advances have been made in understanding the molecular underpinnings of retinal degenerative diseases. Building upon the development of modern techniques for gene delivery, many gene-based therapies have been effectively used to treat loss-of-function retinal diseases in mice and men. Significant effort has been invested into making gene delivery vehicles more efficient, less toxic, and non-immunogenic. However, one challenge for the treatment of more complex gain-of-function diseases, many of which might be benefited by the regulation of cellular stress-responsive signaling pathways, is the ability to control the strategy in a physiological (conditional) manner. This review is focused on promising retinal gene therapy strategies that rely on small molecule-based conditional regulation and the inherent limitations and challenges of these strategies that need to be addressed prior to their extensive use.
Collapse
|
14
|
GDNF-mediated rescue of the nigrostriatal system depends on the degree of degeneration. Gene Ther 2018; 26:57-64. [PMID: 30531868 PMCID: PMC6514883 DOI: 10.1038/s41434-018-0049-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 10/01/2018] [Accepted: 10/12/2018] [Indexed: 02/02/2023]
Abstract
Glial cell-line derived neurotrophic factor (GDNF) is a promising therapeutic molecule to treat Parkinson’s disease. Despite an excellent profile in experimental settings, clinical trials testing GDNF have failed. One of the theories to explain these negative outcomes is that the clinical trials were done in late-stage patients that have advanced nigrostriatal degeneration and may therefore not respond to a neurotrophic factor therapy. Based on this idea, we tested if the stage of nigrostriatal degeneration is important for GDNF-based therapies. Lentiviral vectors expressing regulated GDNF were delivered to the striatum of rats to allow GDNF expression to be turned on either while the nigrostriatal system was degenerating or after the nigrostriatal system had been fully lesioned by 6-OHDA. In the group of animals where GDNF expression was on during degeneration, neurons were rescued and there was a reversal of motor deficits. Turning GDNF expression on after the nigrostriatal system was lesioned did not rescue neurons or reverse motor deficits. In fact, these animals were indistinguishable from the control groups. Our results suggest that GDNF can reverse motor deficits and nigrostriatal pathology despite an ongoing nigrostriatal degeneration, if there is still a sufficient number of remaining neurons to respond to therapy.
Collapse
|
15
|
Huang L, Ozawa M, Miyamoto-Sato E. Development of a novel conditional knockdown mouse based on YB-1 protein degradation. Genes Cells 2018; 23:860-867. [PMID: 30160330 DOI: 10.1111/gtc.12642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 07/04/2018] [Accepted: 07/29/2018] [Indexed: 11/28/2022]
Abstract
To clarify the pathogenic mechanism of disease and establish effective therapies, animal disease models that can be dynamically analyzed are urgently required. Knockout mouse models and conditional genetically engineered mouse models were developed to analyze genes and proteins involved in disease. However, these methods have drawbacks, including embryonic lethality, side effects and low efficiency. To address this issue, we created a novel transgenic mouse model in which the YB1 gene was fused with a destabilizing domain (DD), named the YB1-DD mouse. YB-1 is widely expressed throughout development and has been implicated as a cell survival factor. Newly synthesized DD proteins are degraded through the proteasome pathway, but their degradation can be blocked with trimethoprim (TMP). In this study, we established a novel conditional knockdown mouse model that enables targeting of protein degradation directly; this model resulted in dose-dependent regulation of the target protein YB-1 by the ligand TMP in YB1 heterozygous mice. Since this conditional knockdown mouse model appears to be functional, it has potential as a useful disease model based on direct protein degradation control.
Collapse
Affiliation(s)
- Lijuan Huang
- Division of Molecular Biology, Research Institute for Biomedical Science, Tokyo University of Science, Noda, Japan
| | - Masaaki Ozawa
- Division of Molecular Biology, Research Institute for Biomedical Science, Tokyo University of Science, Noda, Japan
| | - Etsuko Miyamoto-Sato
- Division of Molecular Biology, Research Institute for Biomedical Science, Tokyo University of Science, Noda, Japan
| |
Collapse
|
16
|
Santiago CP, Keuthan CJ, Boye SL, Boye SE, Imam AA, Ash JD. A Drug-Tunable Gene Therapy for Broad-Spectrum Protection against Retinal Degeneration. Mol Ther 2018; 26:2407-2417. [PMID: 30078764 PMCID: PMC6171322 DOI: 10.1016/j.ymthe.2018.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/27/2022] Open
Abstract
Retinal degenerations are a large cluster of diseases characterized by the irreversible loss of light-sensitive photoreceptors that impairs the vision of 9.1 million people in the US. An attractive treatment option is to use gene therapy to deliver broad-spectrum neuroprotective factors. However, this approach has had limited clinical translation because of the inability to control transgene expression. To address this problem, we generated an adeno-associated virus vector named RPF2 that was engineered to express domains of leukemia inhibitory factor fused to the destabilization domain of bacterial dihydrofolate reductase. Fusion proteins containing the destabilization domain are degraded in mammalian cells but can be stabilized with the binding of the drug trimethoprim. Our data show that expression levels of RPF2 are tightly regulated by the dose of trimethoprim and can be reversed by trimethoprim withdrawal. We further show that stabilized RPF2 can protect photoreceptors and prevent blindness in treated mice.
Collapse
Affiliation(s)
- Clayton P Santiago
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Casey J Keuthan
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Aisha A Imam
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - John D Ash
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
17
|
Datta S, Renwick M, Chau VQ, Zhang F, Nettesheim ER, Lipinski DM, Hulleman JD. A Destabilizing Domain Allows for Fast, Noninvasive, Conditional Control of Protein Abundance in the Mouse Eye - Implications for Ocular Gene Therapy. Invest Ophthalmol Vis Sci 2018; 59:4909-4920. [PMID: 30347085 PMCID: PMC6181441 DOI: 10.1167/iovs.18-24987] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/30/2018] [Indexed: 01/23/2023] Open
Abstract
Purpose Temporal and reversible control of protein expression in vivo is a central goal for many gene therapies, especially for strategies involving proteins that are detrimental to physiology if constitutively expressed. Accordingly, we explored whether protein abundance in the mouse retina could be effectively controlled using a destabilizing Escherichia coli dihydrofolate reductase (DHFR) domain whose stability is dependent on the small molecule, trimethoprim (TMP). Methods We intravitreally injected wild-type C57BL6/J mice with an adeno-associated vector (rAAV2/2[MAX]) constitutively expressing separate fluorescent reporters: DHFR fused to yellow fluorescent protein (DHFR.YFP) and mCherry. TMP or vehicle was administered to mice via oral gavage, drinking water, or eye drops. Ocular TMP levels post treatment were quantified by LC-MS/MS. Protein abundance was measured by fundus fluorescence imaging and western blotting. Visual acuity, response to light stimulus, retinal structure, and gene expression were evaluated after long-term (3 months) TMP treatment. Results Without TMP, DHFR.YFP was efficiently degraded in the retina. TMP achieved ocular concentrations of ∼13.6 μM (oral gavage), ∼331 nM (drinking water), and ∼636 nM (eye drops). Oral gavage and TMP eye drops stabilized DHFR.YFP as quickly as 6 hours, whereas continuous TMP drinking water could stabilize DHFR.YFP for ≥3 months. Stabilization was completely and repeatedly reversible following removal/addition of TMP in all regimens. Long-term TMP treatment had no impact on retina function/structure and had no effect on >99.9% of tested genes. Conclusions This DHFR-based conditional system is a rapid, efficient, and reversible tool to effectively control protein expression in the retina.
Collapse
Affiliation(s)
- Shyamtanu Datta
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Marian Renwick
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Viet Q. Chau
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Fang Zhang
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Emily R. Nettesheim
- Department of Ophthalmology, Eye Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Daniel M. Lipinski
- Department of Ophthalmology, Eye Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
| | - John D. Hulleman
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
18
|
Quintino L, Namislo A, Davidsson M, Breger LS, Kavanagh P, Avallone M, Elgstrand-Wettergren E, Isaksson C, Lundberg C. Destabilizing Domains Enable Long-Term and Inert Regulation of GDNF Expression in the Brain. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:29-39. [PMID: 30324128 PMCID: PMC6187056 DOI: 10.1016/j.omtm.2018.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/30/2018] [Indexed: 12/31/2022]
Abstract
Regulation of therapeutic transgene expression can increase the safety of gene therapy interventions, especially when targeting critical organs such as the brain. Although several gene expression systems have been described, none of the current systems has the required safety profile for clinical applications. Our group has previously adapted a system for novel gene regulation based on the destabilizing domain degron technology to successfully regulate glial cell-line derived neurotrophic factor in the brain (GDNF-F-DD). In the present study, we used GDNF-F-DD as a proof-of-principle molecule to fully characterize DD regulation in the brain. Our results indicate that DD could be regulated in a dose-dependent manner. In addition, GDNF-F-DD could also be induced in vivo repeatedly, without loss of activity or efficacy in vivo. Finally, DD regulation was able to be sustained for 24 weeks without loss of expression or any overt toxicity. The present study shows that DD has great potential to regulate gene expression in the brain.
Collapse
Affiliation(s)
- Luis Quintino
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Angrit Namislo
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Marcus Davidsson
- Molecular Neuromodulation, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Ludivine S Breger
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Patrick Kavanagh
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Martino Avallone
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Christina Isaksson
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cecilia Lundberg
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Harrison H, Pegg HJ, Thompson J, Bates C, Shore P. HIF1-alpha expressing cells induce a hypoxic-like response in neighbouring cancer cells. BMC Cancer 2018; 18:674. [PMID: 29925335 PMCID: PMC6011406 DOI: 10.1186/s12885-018-4577-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/05/2018] [Indexed: 12/26/2022] Open
Abstract
Background Hypoxia stimulates metastasis in cancer and is linked to poor patient prognosis. In tumours, oxygen levels vary and hypoxic regions exist within a generally well-oxygenated tumour. However, whilst the heterogeneous environment is known to contribute to metastatic progression, little is known about the mechanism by which heterogeneic hypoxia contributes to cancer progression. This is largely because existing experimental models do not recapitulate the heterogeneous nature of hypoxia. The primary effector of the hypoxic response is the transcription factor Hypoxia inducible factor 1-alpha (HIF1-alpha). HIF1-alpha is stabilised in response to low oxygen levels in the cellular environment and its expression is seen in hypoxic regions throughout the tumour. Methods We have developed a model system in which HIF1-alpha can be induced within a sub-population of cancer cells, thus enabling us to mimic the effects of heterogeneic HIF1-alpha expression. Results We show that induction of HIF1-alpha not only recapitulates elements of the hypoxic response in the induced cells but also results in significant changes in proliferation, gene expression and mammosphere formation within the HIF1-alpha negative population. Conclusions These findings suggest that the HIF1-alpha expressing cells found within hypoxic regions are likely to contribute to the subsequent progression of a tumour by modifying the behaviour of cells in the non-hypoxic regions of the local micro-environment. Electronic supplementary material The online version of this article (10.1186/s12885-018-4577-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hannah Harrison
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| | - Henry J Pegg
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jamie Thompson
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Christian Bates
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Paul Shore
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
20
|
Park S, Burke RE, Kareva T, Kholodilov N, Aimé P, Franke TF, Levy O, Greene LA. Context-dependent expression of a conditionally-inducible form of active Akt. PLoS One 2018; 13:e0197899. [PMID: 29920520 PMCID: PMC6007834 DOI: 10.1371/journal.pone.0197899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
Akt kinases are key signaling components in proliferation-competent and post-mitotic cells. Here, we sought to create a conditionally-inducible form of active Akt for both in vitro and in vivo applications. We fused a ligand-responsive Destabilizing Domain (DD) derived from E. coli dihydrofolate reductase to a constitutively active mutant form of Akt1, Akt(E40K). Prior work indicated that such fusion proteins may be stabilized and induced by a ligand, the antibiotic Trimethoprim (TMP). We observed dose-dependent, reversible induction of both total and phosphorylated/active DD-Akt(E40K) by TMP across several cellular backgrounds in culture, including neurons. Phosphorylation of FoxO4, an Akt substrate, was significantly elevated after DD-Akt(E40K) induction, indicating the induced protein was functionally active. The induced Akt(E40K) protected cells from apoptosis evoked by serum deprivation and was neuroprotective in two cellular models of Parkinson's disease (6-OHDA and MPP+ exposure). There was no significant protection without induction. We also evaluated Akt(E40K) induction by TMP in mouse substantia nigra and striatum after neuronal delivery via an AAV1 adeno-associated viral vector. While there was significant induction in striatum, there was no apparent induction in substantia nigra. To explore the possible basis for this difference, we examined DD-Akt(E40K) induction in cultured ventral midbrain neurons. Both dopaminergic and non-dopaminergic neurons in the cultures showed DD-Akt(E40K) induction after TMP treatment. However, basal DD-Akt(E40K) expression was 3-fold higher for dopaminergic neurons, resulting in a significantly lower induction by TMP in this population. Such findings suggest that dopaminergic neurons may be relatively inefficient in protein degradation, a property that could relate to their lack of apparent DD-Akt(E40K) induction in vivo and to their selective vulnerability in Parkinson's disease. In summary, we generated an inducible, biologically active form of Akt. The degree of inducibility appears to reflect cellular context that will inform the most appropriate applications for this and related reagents.
Collapse
Affiliation(s)
- Soyeon Park
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - Robert E Burke
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America.,Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Tatyana Kareva
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Nikolai Kholodilov
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Pascaline Aimé
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Thomas F Franke
- Department of Neuroscience, Icahn School of Medicine at Mt Sinai, New York, New York, United States of America
| | - Oren Levy
- Department of Neurology, Columbia University Medical Center, New York, New York, United States of America
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
21
|
Czapiński J, Kiełbus M, Kałafut J, Kos M, Stepulak A, Rivero-Müller A. How to Train a Cell-Cutting-Edge Molecular Tools. Front Chem 2017; 5:12. [PMID: 28344971 PMCID: PMC5344921 DOI: 10.3389/fchem.2017.00012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/20/2017] [Indexed: 12/28/2022] Open
Abstract
In biological systems, the formation of molecular complexes is the currency for all cellular processes. Traditionally, functional experimentation was targeted to single molecular players in order to understand its effects in a cell or animal phenotype. In the last few years, we have been experiencing rapid progress in the development of ground-breaking molecular biology tools that affect the metabolic, structural, morphological, and (epi)genetic instructions of cells by chemical, optical (optogenetic) and mechanical inputs. Such precise dissection of cellular processes is not only essential for a better understanding of biological systems, but will also allow us to better diagnose and fix common dysfunctions. Here, we present several of these emerging and innovative techniques by providing the reader with elegant examples on how these tools have been implemented in cells, and, in some cases, organisms, to unravel molecular processes in minute detail. We also discuss their advantages and disadvantages with particular focus on their translation to multicellular organisms for in vivo spatiotemporal regulation. We envision that further developments of these tools will not only help solve the processes of life, but will give rise to novel clinical and industrial applications.
Collapse
Affiliation(s)
- Jakub Czapiński
- Department of Biochemistry and Molecular Biology, Medical University of LublinLublin, Poland
- Postgraduate School of Molecular Medicine, Medical University of WarsawWarsaw, Poland
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of LublinLublin, Poland
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of LublinLublin, Poland
| | - Michał Kos
- Department of Biochemistry and Molecular Biology, Medical University of LublinLublin, Poland
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of LublinLublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of LublinLublin, Poland
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi UniversityTurku, Finland
- Department of Biosciences, Åbo Akademi UniversityTurku, Finland
| |
Collapse
|
22
|
A regulatable AAV vector mediating GDNF biological effects at clinically-approved sub-antimicrobial doxycycline doses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16027. [PMID: 27069954 PMCID: PMC4813607 DOI: 10.1038/mtm.2016.27] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
Abstract
Preclinical and clinical data stress the importance of pharmacologically-controlling glial cell line-derived neurotrophic factor (GDNF) intracerebral administration to treat PD. The main challenge is finding a combination of a genetic switch and a drug which, when administered at a clinically-approved dose, reaches the brain in sufficient amounts to induce a therapeutic effect. We describe a highly-sensitive doxycycline-inducible adeno-associated virus (AAV) vector. This vector allowed for the first time a longitudinal analysis of inducible transgene expression in the brain using bioluminescence imaging. To evaluate the dose range of GDNF biological activity, the inducible AAV vector (8.0 × 10(9) viral genomes) was injected in the rat striatum at four delivery sites and increasing doxycycline doses administered orally. ERK/Akt signaling activation as well as tyrosine hydroxylase downregulation, a consequence of long-term GDNF treatment, were induced at plasmatic doxycycline concentrations of 140 and 320 ng/ml respectively, which are known not to increase antibiotic-resistant microorganisms in patients. In these conditions, GDNF covered the majority of the striatum. No behavioral abnormalities or weight loss were observed. Motor asymmetry resulting from unilateral GDNF treatment only appeared with a 2.5-fold higher vector and a 13-fold higher inducer doses. Our data suggest that using the herein-described inducible AAV vector, biological effects of GDNF can be obtained in response to sub-antimicrobial doxycycline doses.
Collapse
|
23
|
Abstract
Gene therapy represents a promising approach for the treatment of monogenic and multifactorial neurological disorders. It can be used to replace a missing gene and mutated gene or downregulate a causal gene. Despite the versatility of gene therapy, one of the main limitations lies in the irreversibility of the process: once delivered to target cells, the gene of interest is constitutively expressed and cannot be removed. Therefore, efficient, safe and long-term gene modification requires a system allowing fine control of transgene expression.Different systems have been developed over the past decades to regulate transgene expression after in vivo delivery, either at transcriptional or post-translational levels. The purpose of this chapter is to give an overview on current regulatory system used in the context of gene therapy for neurological disorders. Systems using external regulation of transgenes using antibiotics are commonly used to control either gene expression using tetracycline-controlled transcription or protein levels using destabilizing domain technology. Alternatively, specific promoters of genes that are regulated by disease mechanisms, increasing expression as the disease progresses or decreasing expression as disease regresses, are also examined. Overall, this chapter discusses advantages and drawbacks of current molecular methods for regulated gene therapy in the central nervous system.
Collapse
Affiliation(s)
- Ludivine Breger
- Department of Experimental Medical Sciences, CNS Gene Therapy Unit, Wallenberg Neuroscience Center, Lund University, BMC A11, 221 84, Lund, Sweden
| | - Erika Elgstrand Wettergren
- Department of Experimental Medical Sciences, CNS Gene Therapy Unit, Wallenberg Neuroscience Center, Lund University, BMC A11, 221 84, Lund, Sweden
| | - Luis Quintino
- Department of Experimental Medical Sciences, CNS Gene Therapy Unit, Wallenberg Neuroscience Center, Lund University, BMC A11, 221 84, Lund, Sweden
| | - Cecilia Lundberg
- Department of Experimental Medical Sciences, CNS Gene Therapy Unit, Wallenberg Neuroscience Center, Lund University, BMC A11, 221 84, Lund, Sweden.
| |
Collapse
|
24
|
Domanskyi A, Saarma M, Airavaara M. Prospects of Neurotrophic Factors for Parkinson's Disease: Comparison of Protein and Gene Therapy. Hum Gene Ther 2015; 26:550-9. [DOI: 10.1089/hum.2015.065] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Andrii Domanskyi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Froschauer A, Kube L, Kegler A, Rieger C, Gutzeit HO. Tunable Protein Stabilization In Vivo Mediated by Shield-1 in Transgenic Medaka. PLoS One 2015; 10:e0131252. [PMID: 26148066 PMCID: PMC4493054 DOI: 10.1371/journal.pone.0131252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/29/2015] [Indexed: 11/19/2022] Open
Abstract
Techniques for conditional gene or protein expression are important tools in developmental biology and in the analysis of physiology and disease. On the protein level, the tunable and reversible expression of proteins can be achieved by the fusion of the protein of interest to a destabilizing domain (DD). In the absence of its specific ligand (Shield-1), the protein is degraded by the proteasome. The DD-Shield system has proven to be an excellent tool to regulate the expression of proteins of interests in mammalian systems but has not been applied in teleosts like the medaka. We present the application of the DD-Shield technique in transgenic medaka and show the ubiquitous conditional expression throughout life. Shield-1 administration to the water leads to concentration-dependent induction of a YFP reporter gene in various organs and in spermatogonia at the cellular level.
Collapse
Affiliation(s)
| | - Lisa Kube
- Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Alexandra Kegler
- Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Christiane Rieger
- Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Herwig O. Gutzeit
- Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
26
|
Saayman S, Ali SA, Morris KV, Weinberg MS. The therapeutic application of CRISPR/Cas9 technologies for HIV. Expert Opin Biol Ther 2015; 15:819-30. [PMID: 25865334 DOI: 10.1517/14712598.2015.1036736] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The use of antiretroviral therapy has led to a significant decrease in morbidity and mortality in HIV-infected individuals. Nevertheless, gene-based therapies represent a promising therapeutic paradigm for HIV-1, as they have the potential for sustained viral inhibition and reduced treatment interventions. One new method amendable to a gene-based therapy is the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein-9 nuclease (Cas9) gene editing system. AREAS COVERED CRISPR/Cas9 can be engineered to successfully modulate an array of disease-causing genetic elements. We discuss the diverse roles that CRISPR/Cas9 may play in targeting HIV and eradicating infection. The Cas9 nuclease coupled with one or more small guide RNAs can target the provirus to mediate excision of the integrated viral genome. Moreover, a modified nuclease-deficient Cas9 fused to transcription activation domains may induce targeted activation of proviral gene expression allowing for the purging of the latent reservoirs. These technologies can also be exploited to target host dependency factors such as the co-receptor CCR5, thus preventing cellular entry of the virus. EXPERT OPINION The diversity of the CRISPR/Cas9 technologies offers great promise for targeting different stages of the viral life cycle, and have the capacity for mediating an effective and sustained genetic therapy against HIV.
Collapse
Affiliation(s)
- Sheena Saayman
- The Scripps Research Institute, Department of Molecular and Experimental Medicine , 10550 North Torrey Pines Road, La Jolla, CA, 92037 , USA
| | | | | | | |
Collapse
|
27
|
d'Anglemont de Tassigny X, Pascual A, López-Barneo J. GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway. Implications for Parkinson's disease. Front Neuroanat 2015; 9:10. [PMID: 25762899 PMCID: PMC4327623 DOI: 10.3389/fnana.2015.00010] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
The glial cell line-derived neurotrophic factor (GDNF) is a well-established trophic agent for dopaminergic (DA) neurons in vitro and in vivo. GDNF is necessary for maintenance of neuronal morphological and neurochemical phenotype and protects DA neurons from toxic damage. Numerous studies on animal models of Parkinson’s disease (PD) have reported beneficial effects of GDNF on nigrostriatal DA neuron survival. However, translation of these observations to the clinical setting has been hampered so far by side effects associated with the chronic continuous intra-striatal infusion of recombinant GDNF. In addition, double blind and placebo-controlled clinical trials have not reported any clinically relevant effect of GDNF on PD patients. In the past few years, experiments with conditional Gdnf knockout mice have suggested that GDNF is necessary for maintenance of DA neurons in adulthood. In parallel, new methodologies for exogenous GDNF delivery have been developed. Recently, it has been shown that a small population of scattered, electrically interconnected, parvalbumin positive (PV+) GABAergic interneurons is responsible for most of the GDNF produced in the rodent striatum. In addition, cholinergic striatal interneurons appear to be also involved in the modulation of striatal GDNF. In this review, we summarize current knowledge on brain GDNF delivery, homeostasis, and its effects on nigrostriatal DA neurons. Special attention is paid to the therapeutic potential of endogenous GDNF stimulation in PD.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain ; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla Seville, Spain ; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid, Spain
| |
Collapse
|
28
|
Controlled Striatal DOPA Production From a Gene Delivery System in a Rodent Model of Parkinson's Disease. Mol Ther 2015; 23:896-906. [PMID: 25592335 DOI: 10.1038/mt.2015.8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/29/2014] [Indexed: 12/25/2022] Open
Abstract
Conventional symptomatic treatment for Parkinson's disease (PD) with long-term L-3,4-dihydroxyphenylalanine (DOPA) is complicated with development of drug-induced side effects. In vivo viral vector-mediated gene expression encoding tyrosine hydroxylase (TH) and GTP cyclohydrolase 1 (GCH1) provides a drug delivery strategy of DOPA with distinct advantages over pharmacotherapy. Since the brain alterations made with current gene transfer techniques are irreversible, the therapeutic approaches taken to the clinic should preferably be controllable to match the needs of each individual during the course of their disease. We used a recently described tunable gene expression system based on the use of destabilized dihydrofolate reductase (DD) and generated a N-terminally coupled GCH1 enzyme (DD-GCH1) while the TH enzyme was constitutively expressed, packaged in adeno-associated viral (AAV) vectors. Expression of DD-GCH1 was regulated by the activating ligand trimethoprim (TMP) that crosses the blood-brain barrier. We show that the resulting intervention provides a TMP-dose-dependent regulation of DOPA synthesis that is closely linked to the magnitude of functional effects. Our data constitutes the first proof of principle for controlled reconstitution of dopamine capacity in the brain and suggests that such next-generation gene therapy strategies are now mature for preclinical development toward use in patients with PD.
Collapse
|
29
|
Tereshchenko J, Maddalena A, Bähr M, Kügler S. Pharmacologically controlled, discontinuous GDNF gene therapy restores motor function in a rat model of Parkinson's disease. Neurobiol Dis 2014; 65:35-42. [PMID: 24440408 DOI: 10.1016/j.nbd.2014.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/13/2013] [Accepted: 01/08/2014] [Indexed: 11/18/2022] Open
Abstract
Neurotrophic factors have raised hopes to be able to cure symptoms and to prevent progressive neurodegeneration in devastating neurological diseases. Gene therapy by means of viral vectors can overcome the hurdle of targeted delivery, but its current configuration is irreversible and thus much less controllable than that of classical pharmacotherapies. We thus aimed at developing a strategy allowing for both curative and controllable neurotrophic factor expression. Therefore, the short-term, intermittent and reversible expression of a neutrophic factor was evaluated for therapeutic efficacy in a slowly progressive animal model of Parkinson's disease (PD). We demonstrate that short-term induced expression of glial cell line derived neurotrophic factor (GDNF) is sufficient to provide i) substantial protection of nigral dopaminergic neurons from degeneration and ii) restoration of dopamine supply and motor behaviour in the partial striatal 6-OHDA model PD. These neurorestorative effects of GDNF lasted several weeks beyond the time of its expression. Later on, therapeutic efficacy ceased, but was restored by a second short induction of GDNF expression, demonstrating that monthly application of the inducing drug mifepristone was sufficient to maintain neuroprotective and neurorestorative GDNF levels. These findings suggest that forthcoming gene therapies for PD or other neurodegenerative disorders can be designed in a way that low frequency application of an approved drug can provide controllable and therapeutically efficient levels of GDNF or other neurotrophic factors. Neurotrophic factor expression can be withdrawn in case of off-target effects or sufficient clinical benefit, a feature that may eventually increase the acceptance of gene therapy for less advanced patients, which may profit better from such approaches.
Collapse
Affiliation(s)
- Julia Tereshchenko
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Andrea Maddalena
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Mathias Bähr
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Sebastian Kügler
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany.
| |
Collapse
|
30
|
Seijsing J, Lindborg M, Löfblom J, Uhlén M, Gräslund T. Robust expression of the human neonatal Fc receptor in a truncated soluble form and as a full-length membrane-bound protein in fusion with eGFP. PLoS One 2013; 8:e81350. [PMID: 24260574 PMCID: PMC3832405 DOI: 10.1371/journal.pone.0081350] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 10/11/2013] [Indexed: 11/20/2022] Open
Abstract
Studies on the neonatal Fc receptor (FcRn) have revealed a multitude of important functions in mammals, including protection of IgG and serum albumin (SA) from lysosomal degradation. The pharmacokinetic behavior of therapeutic antibodies, IgG-Fc- and SA-containing drugs is therefore influenced by their interaction with FcRn. Pre-clinical development of such drugs is facilitated if their interaction with FcRn can be studied in vitro. For this reason we have developed a robust system for production of the soluble extracellular domain of human FcRn as well as the full-length receptor as fusion to green fluorescent protein, taking advantage of a lentivirus-based gene delivery system where stable over-expressing cells are easily and rapidly generated. Production of the extracellular domain in multiple-layered culture flasks, followed by affinity purification using immobilized IgG, resulted in capture of milligram amounts of soluble receptor per liter cell culture with retained IgG binding. The receptor was further characterized by SDS-PAGE, western blotting, circular dichroism spectroscopy, ELISA, surface plasmon resonance and a temperature stability assay showing a functional and stable protein of high purity. The full-length receptor was found to be successfully over-expressed in a membrane-bound form with retained pH-dependent IgG- and SA-binding.
Collapse
Affiliation(s)
- Johan Seijsing
- School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - John Löfblom
- School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Torbjörn Gräslund
- School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
31
|
Functional neuroprotection and efficient regulation of GDNF using destabilizing domains in a rodent model of Parkinson's disease. Mol Ther 2013; 21:2169-80. [PMID: 23881415 DOI: 10.1038/mt.2013.169] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 07/10/2013] [Indexed: 11/08/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has great potential to treat Parkinson's disease (PD). However, constitutive expression of GDNF can over time lead to side effects. Therefore, it would be useful to regulate GDNF expression. Recently, a new gene inducible system using destabilizing domains (DD) from E. coli dihydrofolate reductase (DHFR) has been developed and characterized. The advantage of this novel DD is that it is regulated by trimethoprim (TMP), a well-characterized drug that crosses the blood-brain barrier and can therefore be used to regulate gene expression in the brain. We have adapted this system to regulate expression of GDNF. A C-terminal fusion of GDNF and a DD with an additional furin cleavage site was able to be efficiently regulated in vitro, properly processed and was able to bind to canonical GDNF receptors, inducing a signaling cascade response in target cells. In vivo characterization of the protein showed that it could be efficiently induced by TMP and it was only functional when gene expression was turned on. Further characterization in a rodent model of PD showed that the regulated GDNF protected neurons, improved motor behavior of animals and was efficiently regulated in a pathological setting.
Collapse
|
32
|
Quintino L, Baudet A, Larsson J, Lundberg C. FACS binding assay for analysing GDNF interactions. J Neurosci Methods 2013; 218:25-8. [PMID: 23669067 DOI: 10.1016/j.jneumeth.2013.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 11/29/2022]
Abstract
Glial cell-line derived neurotrophic factor (GDNF) is a secreted protein with great therapeutic potential. However, in order to analyse the interactions between GDNF and its receptors, researchers have been mostly dependent of radioactive binding assays. We developed a FACS-based binding assay for GDNF as an alternative to current methods. We demonstrated that the FACS-based assay using TGW cells allowed readily detection of GDNF binding and displacement to endogenous receptors. The dissociation constant and half maximal inhibitory concentration obtained were comparable to other studies using standard binding assays. Overall, this FACS-based, simple to perform and adaptable to high throughput setup, provides a safer and reliable alternative to radioactive methods.
Collapse
Affiliation(s)
- Luís Quintino
- CNS Gene Therapy, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|