1
|
Soltysiak MPM, Ory ALH, Lee AD, Christophersen CE, Jalihal AP, Springer M. XanthoMoClo─A Robust Modular Cloning Genetic Toolkit for the Genera Xanthobacter and Roseixanthobacter. ACS Synth Biol 2025; 14:1173-1190. [PMID: 40080684 PMCID: PMC12012871 DOI: 10.1021/acssynbio.4c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/27/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Interest in Xanthobacter species is increasing due to their unique metabolic capabilities. They can grow in both heterotrophic and fully autotrophic environments, including carbon dioxide, dinitrogen gas, and hydrogen as the sole carbon, nitrogen, and energy sources, respectively. Academic and industrial groups looking to leverage these metabolic properties are already using Xanthobacter strains for the sustainable production of food and commodities. However, only a handful of genetic parts and protocols exist in scattered genetic backgrounds, and there is an unmet need for reliable genetic engineering tools to manipulate Xanthobacter species. Here, we developed XanthoMoClo, a robust modular cloning genetic toolkit for Xanthobacter and Roseixanthobacter species and strains, providing extensive tools to transform them, manipulate their metabolism, and express genes of interest. The toolkit contains plasmid parts, such as replication origins, antibiotic selection markers, fluorescent proteins, constitutive and inducible promoters, a standardized framework to incorporate novel components into the toolkit, and a conjugation donor to transform Xanthobacter and Roseixanthobacter strains easily with no or minimal optimization. We validated these plasmid components in depth in three of the most commonly studied Xanthobacter strains: X. versatilis Py2, X. autotrophicus GZ29, and X. flavus GJ10, as well as in R. finlandensis VTT E-85241. Finally, we demonstrate robust toolkit functionality across 21 different species of Xanthobacter and Roseixanthobacter, comprising 23 strains in total. The XanthoMoClo genetic toolkit is available to the research community (through AddGene) and will help accelerate the genetic engineering of Xanthobacter to further their applications in sustainability and bioremediation efforts.
Collapse
Affiliation(s)
| | - Audrey L. H. Ory
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Andrew D. Lee
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | | | - Amogh P. Jalihal
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Michael Springer
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Broad
Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, United States
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
2
|
Balmer GL, Guha S, Poll S. Engrams across diseases: Different pathologies - unifying mechanisms? Neurobiol Learn Mem 2025; 219:108036. [PMID: 40023216 DOI: 10.1016/j.nlm.2025.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Memories are our reservoir of knowledge and thus, are crucial for guiding decisions and defining our self. The physical correlate of a memory in the brain is termed an engram and since decades helps researchers to elucidate the intricate nature of our imprinted experiences and knowledge. Given the importance that memories have for our lives, their impairment can present a tremendous burden. In this review we aim to discuss engram malfunctioning across diseases, covering dementia-associated pathologies, epilepsy, chronic pain and psychiatric disorders. Current neuroscientific tools allow to witness the emergence and fate of engram cells and enable their manipulation. We further suggest that specific mechanisms of mnemonic malfunction can be derived from engram cell readouts. While depicting the way diseases act on the mnemonic component - specifically, on the cellular engram - we emphasize a differentiation between forms of amnesia and hypermnesia. Finally, we highlight commonalities and distinctions of engram impairments on the cellular level across diseases independent of their pathogenic origins and discuss prospective therapeutic measures.
Collapse
Affiliation(s)
- Greta Leonore Balmer
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Shuvrangshu Guha
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Stefanie Poll
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE) Bonn, Germany.
| |
Collapse
|
3
|
Abidi S, Elhazaz Fernandez A, Seehase N, Hanisch L, Karlas A, Sandig V, Jordan I. Expression of an Efficient Selection Marker Out of a Duplicated Site in the ITRs of a Modified Vaccinia Virus Ankara (MVA). Vaccines (Basel) 2024; 12:1377. [PMID: 39772039 PMCID: PMC11680203 DOI: 10.3390/vaccines12121377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Poxviruses are large DNA viruses that replicate in the host cytoplasm without a nuclear phase. As vaccine vectors, they can package and express large recombinant cassettes from different positions of their genomic core region. We present a comparison between wildtype modified vaccinia Ankara (MVA) and isolate CR19, which has significantly expanded inverted terminal repeats (ITRs). With this expansion, a site in wildtype MVA, called deletion site (DS) IV, has been duplicated at both ends of the genome and now occupies an almost central position in the newly formed ITRs. Methods: We inserted various reporter genes into this site and found that the ITRs can be used for transgene expression. However, ITRs are genomic structures that can rapidly adapt to selective pressure through transient duplication and contraction. To test the potential utility of insertions into viral telomers, we inserted a factor from the cellular innate immune system that interferes with viral replication as an example of a difficult transgene. Results: A site almost in the centre of the ITRs can be used for transgene expression, and both sides are mirrored into identical copies. The example of a challenging transgene, tetherin, proved to be surprisingly efficient in selecting candidate vectors against the large background of parental viruses. Conclusions: Insertion of transgenes into ITRs automatically doubles the gene doses. The functionalisation of viruses with tetherin may accelerate the identification and generation of recombinant vectors for personalised medicine and pandemic preparedness.
Collapse
Affiliation(s)
- Sirine Abidi
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
| | - Aurora Elhazaz Fernandez
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
- Berlin Institute for Medical Systems Biology (BIMSB), 10115 Berlin, Germany
| | - Nicole Seehase
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
- Tentamus Pharma & Med Deutschland GmbH, 76149 Karlsruhe, Germany
| | - Lina Hanisch
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
| | - Alexander Karlas
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
| | - Volker Sandig
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
| | - Ingo Jordan
- ProBioGen AG, 13086 Berlin, Germany; (S.A.); (A.E.F.); (N.S.); (A.K.); (V.S.)
| |
Collapse
|
4
|
Wu GY, Zheng XX, Zhao SL, Wang Y, Jiang S, Wang YS, Yi YL, Yao J, Wen HZ, Liu J, Li HL, Sui JF. The prelimbic cortex regulates itch processing by controlling attentional bias. iScience 2022; 26:105829. [PMID: 36619983 PMCID: PMC9816985 DOI: 10.1016/j.isci.2022.105829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/31/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Itch is a complex and unpleasant sensory experience. Recent studies have begun to investigate the neural mechanisms underlying the modulation of sensory and emotional components of itch in the brain. However, the key brain regions and neural mechanism involved in modulating the attentional processing of itch remain elusive. Here, we showed that the prelimbic cortex (PrL) is associated with itch processing and that the manipulation of itch-responsive neurons in the PrL significantly disrupted itch-induced scratching. Interestingly, we found that increasing attentional bias toward a distracting stimulus could disturb itch processing. We also demonstrated the existence of a population of attention-related neurons in the PrL that drive attentional bias to regulate itch processing. Importantly, itch-responsive neurons and attention-related neurons significantly overlapped in the PrL and were mutually interchangeable in the regulation of itch processing at the cellular activity level. Our results revealed that the PrL regulates itch processing by controlling attentional bias.
Collapse
Affiliation(s)
- Guang-Yan Wu
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Corresponding author
| | - Xiao-Xia Zheng
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Shan-Lan Zhao
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi Wang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Shan Jiang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi-Song Wang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi-Lun Yi
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Juan Yao
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hui-Zhong Wen
- Department of Neurobiology, College of Basic Medical Sciences, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Ju Liu
- Department of Foreign Languages, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hong-Li Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Corresponding author
| | - Jian-Feng Sui
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Corresponding author
| |
Collapse
|
5
|
Ortega-de San Luis C, Ryan TJ. Understanding the physical basis of memory: Molecular mechanisms of the engram. J Biol Chem 2022; 298:101866. [PMID: 35346687 PMCID: PMC9065729 DOI: 10.1016/j.jbc.2022.101866] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/18/2022] Open
Abstract
Memory, defined as the storage and use of learned information in the brain, is necessary to modulate behavior and critical for animals to adapt to their environments and survive. Despite being a cornerstone of brain function, questions surrounding the molecular and cellular mechanisms of how information is encoded, stored, and recalled remain largely unanswered. One widely held theory is that an engram is formed by a group of neurons that are active during learning, which undergoes biochemical and physical changes to store information in a stable state, and that are later reactivated during recall of the memory. In the past decade, the development of engram labeling methodologies has proven useful to investigate the biology of memory at the molecular and cellular levels. Engram technology allows the study of individual memories associated with particular experiences and their evolution over time, with enough experimental resolution to discriminate between different memory processes: learning (encoding), consolidation (the passage from short-term to long-term memories), and storage (the maintenance of memory in the brain). Here, we review the current understanding of memory formation at a molecular and cellular level by focusing on insights provided using engram technology.
Collapse
Affiliation(s)
- Clara Ortega-de San Luis
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
| | - Tomás J Ryan
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Franceschini A, Costantini I, Pavone FS, Silvestri L. Dissecting Neuronal Activation on a Brain-Wide Scale With Immediate Early Genes. Front Neurosci 2020; 14:569517. [PMID: 33192255 PMCID: PMC7645181 DOI: 10.3389/fnins.2020.569517] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Visualizing neuronal activation on a brain-wide scale yet with cellular resolution is a fundamental technical challenge for neuroscience. This would enable analyzing how different neuronal circuits are disrupted in pathology and how they could be rescued by pharmacological treatments. Although this goal would have appeared visionary a decade ago, recent technological advances make it eventually feasible. Here, we review the latest developments in the fields of genetics, sample preparation, imaging, and image analysis that could be combined to afford whole-brain cell-resolution activation mapping. We show how the different biochemical and optical methods have been coupled to study neuronal circuits at different spatial and temporal scales, and with cell-type specificity. The inventory of techniques presented here could be useful to find the tools best suited for a specific experiment. We envision that in the next years, mapping of neuronal activation could become routine in many laboratories, allowing dissecting the neuronal counterpart of behavior.
Collapse
Affiliation(s)
| | - Irene Costantini
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy
| | - Francesco S Pavone
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy.,Department of Physics and Astronomy, University of Florence, Florence, Italy
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy.,Department of Physics and Astronomy, University of Florence, Florence, Italy
| |
Collapse
|
7
|
Mepham TB, Combes RD, Balls M, Barbieri O, Blokhuis HJ, Costa P, Crilly RE, de Cock Buning T, Delpire VC, O'Hare MJ, Houdebine LM, van Kreijl CF, van der Meer M, Reinhardt CA, Wolf E, van Zeller AM. The Use of Transgenic Animals in the European Union. Altern Lab Anim 2020. [DOI: 10.1177/026119299802600108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- T. Ben Mepham
- ECVAM, JRC Environment Institute, 21020 Ispra (VA), Italy
| | - Robert D. Combes
- Dipartimento di Oncologia Clinica e Sperimentale, Università di Genova, IST/CBA, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Michael Balls
- Institute for Animal Science and Health (ID-DL), Department of Behaviour, Stress Physiology and Management, Edelhertweg 15, 8200 AB Lelystad, The Netherlands
| | - Ottavia Barbieri
- Instituto di Biologia Molecolare, Via Pontina KM 30.600, 00040 Pomezia, Rome, Italy
| | - Harry J. Blokhuis
- Department for the Study of Animal Experiments, University of Leiden, 2301 CB Leiden, The Netherlands
| | - Patrizia Costa
- Breast Cancer Laboratory, LICR/UCL, 67–73 Riding House Street, London W1P 7LD, UK
| | | | - Tjard de Cock Buning
- Laboratoire de Biologie Cellulaire et Moleculaire, Institut National de la Recherche Agronomique, Domaine de Vilvert, 78352 Jouy-en-Josas, France
| | - Véronique C. Delpire
- Institute for Animal Science and Health (ID-DL), Department of Behaviour, Stress Physiology and Management, Edelhertweg 15, 8200 AB Lelystad, The Netherlands
| | | | - Louis-Marie Houdebine
- Laboratoire de Biologie Cellulaire et Moleculaire, Institut National de la Recherche Agronomique, Domaine de Vilvert, 78352 Jouy-en-Josas, France
| | | | - Miriam van der Meer
- Department of Laboratory Animal Science, Utrecht University, 3508 TD Utrecht, The Netherlands
| | | | - Eckhard Wolf
- Lehrstuhl für Molekulare Tierzucht, Feodor-Lynen-Strasse 25, 81377 Munich, Germany
| | - Anne-Marie van Zeller
- Dipartimento di Oncologia Clinica e Sperimentale, Università di Genova, IST/CBA, Largo R. Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
8
|
Mepham TB, Combes RD, Balls M, Barbieri O, Blokhuis HJ, Costa P, Crilly RE, de Cock Buning T, Delpire VC, O'Hare MJ, Houdebine LM, van Kreijl CF, van der Meer M, Reinhardt CA, Wolf E, van Zeller AM. The Use of Transgenic Animals in the European Union. Altern Lab Anim 2019. [DOI: 10.1177/026119299902701s02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- T. Ben Mepham
- Centre for Applied Bioethics, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Robert D. Combes
- FRAME, Russell & Burch House, 96–98 North Sherwood Street, Nottingham, NG1 4EE, UK
| | - Michael Balls
- ECVAM, JRC Environment Institute, 21020 Ispra (VA), Italy
| | - Ottavia Barbieri
- Dipartimento di Oncologia Clinica e Sperimentale, Università di Genova, IST/CBA, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Harry J. Blokhuis
- Institute for Animal Science and Health (ID-DL), Department of Behaviour, Stress Physiology and Management, Edelhertweg 15, 8200 AB Lelystad, The Netherlands
| | - Patrizia Costa
- Instituto di Biologia Molecolare, Via Pontina KM 30.600, 00040 Pomezia, Rome, Italy
| | - Robert E. Crilly
- Centre for Applied Bioethics, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Tjard de Cock Buning
- Department for the Study of Animal Experiments, University of Leiden, 2301 CB Leiden, The Netherlands
| | | | - Michael J. O'Hare
- Breast Cancer Laboratory, LICR/UCL, 67–73 Riding House Street, London W1P 7LD, UK
| | - Louis-Marie Houdebine
- Laboratoire de Biologie Cellulaire et Moleculaire, Institut National de la Recherche Agronomique, Domaine de Vilvert, 78352 Jouy-en-Josas, France
| | | | - Miriam van der Meer
- Department of Laboratory Animal Science, Utrecht University, 3508 TD Utrecht, The Netherlands
| | | | - Eckhard Wolf
- Lehrstuhl für Molekulare Tierzucht, Feodor-Lynen-Strasse 25, 81377 Munich, Germany
| | | |
Collapse
|
9
|
Smith C, Frolinger T, Brathwaite J, Sims S, Pasinetti GM. Dietary polyphenols enhance optogenetic recall of fear memory in hippocampal dentate gyrus granule neuron subpopulations. Commun Biol 2018; 1:42. [PMID: 30271926 PMCID: PMC6123622 DOI: 10.1038/s42003-018-0043-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/30/2018] [Indexed: 12/23/2022] Open
Abstract
Grape-derived polyphenols have been investigated for their role in promoting memory in model systems of stress, but little is known about select subpopulations of neurons that are influenced by polyphenols to improve memory performance. Granule neurons in the hippocampal dentate gyrus are vulnerable to stressors that impair contextual memory function and can be influenced by dietary polyphenols. We utilized a c-fos-tTA/TRE-ChR2 optogenetics model in which neurons activated during fear learning are labeled with ChR2-mCherry and can be optically reactivated in a different context to recapitulate the behavioral output of a related memory. Treatment with dietary polyphenols increased fear memory recall and ChR2-mCherry expression in dentate gyrus neurons, suggesting that dietary polyphenols promote recruitment of neurons to a fear memory engram. We show that dietary polyphenols promote memory function and offer a general method to map cellular subpopulations influenced by dietary polyphenols, in part through the mechanism of c-Fos expression enhancement. Chad Smith et al. show that dietary polyphenols, compounds found in grapes, enable mice to remember fearful events more effectively and map this function to the hippocampal dentate gyrus neurons. This study offers a way to identify the cellular subpopulations regulated by dietary polyphenols.
Collapse
Affiliation(s)
- Chad Smith
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,JJ Peters VA Medical Center, Bronx, 10468, VA, USA
| | - Tal Frolinger
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,JJ Peters VA Medical Center, Bronx, 10468, VA, USA
| | - Justin Brathwaite
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,JJ Peters VA Medical Center, Bronx, 10468, VA, USA
| | - Steven Sims
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,JJ Peters VA Medical Center, Bronx, 10468, VA, USA
| | - Giulio M Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA. .,JJ Peters VA Medical Center, Bronx, 10468, VA, USA.
| |
Collapse
|
10
|
Life-Long Genetic and Functional Access to Neural Circuits Using Self-Inactivating Rabies Virus. Cell 2017; 170:382-392.e14. [PMID: 28689641 PMCID: PMC5509544 DOI: 10.1016/j.cell.2017.06.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/26/2017] [Accepted: 06/08/2017] [Indexed: 12/05/2022]
Abstract
Neural networks are emerging as the fundamental computational unit of the brain and it is becoming progressively clearer that network dysfunction is at the core of a number of psychiatric and neurodegenerative disorders. Yet, our ability to target specific networks for functional or genetic manipulations remains limited. Monosynaptically restricted rabies virus facilitates the anatomical investigation of neural circuits. However, the inherent cytotoxicity of the rabies largely prevents its implementation in long-term functional studies and the genetic manipulation of neural networks. To overcome this limitation, we developed a self-inactivating ΔG-rabies virus (SiR) that transcriptionally disappears from the infected neurons while leaving permanent genetic access to the traced network. SiR provides a virtually unlimited temporal window for the study of network dynamics and for the genetic and functional manipulation of neural circuits in vivo without adverse effects on neuronal physiology and circuit function. Self-inactivating rabies virus (SiR) provides life-long access to neural networks SiR has no adverse effects on neuronal physiology and network computations SiR allows open-ended Ca2+ imaging of network activity and functional intervention SiR allows generation of circuit-specific knockouts
Collapse
|
11
|
Amaral FE, Parker D, Randis TM, Kulkarni R, Prince AS, Shirasu-Hiza MM, Ratner AJ. Rational manipulation of mRNA folding free energy allows rheostat control of pneumolysin production by Streptococcus pneumoniae. PLoS One 2015; 10:e0119823. [PMID: 25798590 PMCID: PMC4370707 DOI: 10.1371/journal.pone.0119823] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/16/2015] [Indexed: 12/04/2022] Open
Abstract
The contribution of specific factors to bacterial virulence is generally investigated through creation of genetic “knockouts” that are then compared to wild-type strains or complemented mutants. This paradigm is useful to understand the effect of presence vs. absence of a specific gene product but cannot account for concentration-dependent effects, such as may occur with some bacterial toxins. In order to assess threshold and dose-response effects of virulence factors, robust systems for tunable expression are required. Recent evidence suggests that the folding free energy (ΔG) of the 5’ end of mRNA transcripts can have a significant effect on translation efficiency and overall protein abundance. Here we demonstrate that rational alteration of 5’ mRNA folding free energy by introduction of synonymous mutations allows for predictable changes in pneumolysin (PLY) expression by Streptococcus pneumoniae without the need for chemical inducers or heterologous promoters. We created a panel of isogenic S. pneumoniae strains, differing only in synonymous (silent) mutations at the 5’ end of the PLY mRNA that are predicted to alter ΔG. Such manipulation allows rheostat-like control of PLY production and alters the cytotoxicity of whole S. pneumoniae on primary and immortalized human cells. These studies provide proof-of-principle for further investigation of mRNA ΔG manipulation as a tool in studies of bacterial pathogenesis.
Collapse
Affiliation(s)
- Fábio E. Amaral
- Department of Pediatrics, Columbia University, New York, NY United States of America
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Dane Parker
- Department of Pediatrics, Columbia University, New York, NY United States of America
| | - Tara M. Randis
- Department of Pediatrics, Columbia University, New York, NY United States of America
| | - Ritwij Kulkarni
- Department of Pediatrics, Columbia University, New York, NY United States of America
| | - Alice S. Prince
- Department of Pediatrics, Columbia University, New York, NY United States of America
- Department of Pharmacology, Columbia University, New York, NY, United States of America
| | - Mimi M. Shirasu-Hiza
- Department of Genetics & Development, Columbia University, New York, NY, United States of America
| | - Adam J. Ratner
- Department of Pediatrics, Columbia University, New York, NY United States of America
- * E-mail:
| |
Collapse
|
12
|
Liu X, Ramirez S, Tonegawa S. Inception of a false memory by optogenetic manipulation of a hippocampal memory engram. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130142. [PMID: 24298144 DOI: 10.1098/rstb.2013.0142] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Memories can be easily distorted, and a lack of relevant animal models has largely hindered our understanding of false-memory formation. Here, we first identified a population of cells in the dentate gyrus (DG) of the hippocampus that bear the engrams for a specific context; these cells were naturally activated during the encoding phase of fear conditioning and their artificial reactivation using optogenetics in an unrelated context was sufficient for inducing the fear memory specific to the conditioned context. In a further study, DG or CA1 neurons activated by exposure to a particular context were labelled with channelrhodopsin-2 (ChR2). These neurons were later optically reactivated during fear conditioning in a different context. The DG experimental group showed increased freezing in the original context in which a foot shock was never delivered. The recall of this false memory was context specific, activated similar downstream regions engaged during natural fear-memory recall, and was also capable of driving an active fear response. Together, our data demonstrate that by substituting a natural conditioned stimulus with optogenetically reactivated DG cells that bear contextual memory engrams, it is possible to incept an internally and behaviourally represented false fear memory.
Collapse
Affiliation(s)
- Xu Liu
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Howard Hughes Medical Institute, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, , Cambridge, MA 02139, USA
| | | | | |
Collapse
|
13
|
Liu X, Ramirez S, Pang PT, Puryear CB, Govindarajan A, Deisseroth K, Tonegawa S. Optogenetic stimulation of a hippocampal engram activates fear memory recall. Nature 2012; 484:381-5. [PMID: 22441246 PMCID: PMC3331914 DOI: 10.1038/nature11028] [Citation(s) in RCA: 1069] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 03/15/2012] [Indexed: 12/29/2022]
Abstract
A specific memory is thought to be encoded by a sparse population of neurons. These neurons can be tagged during learning for subsequent identification and manipulation. Moreover, their ablation or inactivation results in reduced memory expression, suggesting their necessity in mnemonic processes. However, the question of sufficiency remains: it is unclear whether it is possible to elicit the behavioural output of a specific memory by directly activating a population of neurons that was active during learning. Here we show in mice that optogenetic reactivation of hippocampal neurons activated during fear conditioning is sufficient to induce freezing behaviour. We labelled a population of hippocampal dentate gyrus neurons activated during fear learning with channelrhodopsin-2 (ChR2) and later optically reactivated these neurons in a different context. The mice showed increased freezing only upon light stimulation, indicating light-induced fear memory recall. This freezing was not detected in non-fear-conditioned mice expressing ChR2 in a similar proportion of cells, nor in fear-conditioned mice with cells labelled by enhanced yellow fluorescent protein instead of ChR2. Finally, activation of cells labelled in a context not associated with fear did not evoke freezing in mice that were previously fear conditioned in a different context, suggesting that light-induced fear memory recall is context specific. Together, our findings indicate that activating a sparse but specific ensemble of hippocampal neurons that contribute to a memory engram is sufficient for the recall of that memory. Moreover, our experimental approach offers a general method of mapping cellular populations bearing memory engrams.
Collapse
Affiliation(s)
- Xu Liu
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Couturier C, Deprez B. Setting Up a Bioluminescence Resonance Energy Transfer High throughput Screening Assay to Search for Protein/Protein Interaction Inhibitors in Mammalian Cells. Front Endocrinol (Lausanne) 2012; 3:100. [PMID: 22973258 PMCID: PMC3438444 DOI: 10.3389/fendo.2012.00100] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022] Open
Abstract
Each step of the cell life and its response or adaptation to its environment are mediated by a network of protein/protein interactions termed "interactome." Our knowledge of this network keeps growing due to the development of sensitive techniques devoted to study these interactions. The bioluminescence resonance energy transfer (BRET) technique was primarily developed to allow the dynamic monitoring of protein/protein interactions (PPI) in living cells, and has widely been used to study receptor activation by intra- or extra-molecular conformational changes within receptors and activated complexes in mammal cells. Some interactions are described as crucial in human pathological processes, and a new class of drugs targeting them has recently emerged. The BRET method is well suited to identify inhibitors of PPI and here is described why and how to set up and optimize a high throughput screening assay based on BRET to search for such inhibitory compounds. The different parameters to take into account when developing such BRET assays in mammal cells are reviewed to give general guidelines: considerations on the targeted interaction, choice of BRET version, inducibility of the interaction, kinetic of the monitored interaction, and of the BRET reading, influence of substrate concentration, number of cells and medium composition used on the Z' factor, and expected interferences from colored or fluorescent compounds.
Collapse
Affiliation(s)
- Cyril Couturier
- Univ Lille Nord de FranceLille, France
- INSERM U761, Biostructures and Drug DiscoveryLille, France
- Université du Droit et de la Santé de LilleLille, France
- Institut Pasteur LilleLille, France
- Pôle de Recherche Interdisciplinaire sur le MédicamentLille, France
- *Correspondence: Cyril Couturier, UMR 761, Biostructure and Drug Discovery, Institut Pasteur de Lille, Université Lille 2, 1 rue du Pr Calmette, 59000 Lille, France. e-mail:
| | - Benoit Deprez
- Univ Lille Nord de FranceLille, France
- INSERM U761, Biostructures and Drug DiscoveryLille, France
- Université du Droit et de la Santé de LilleLille, France
- Institut Pasteur LilleLille, France
- Pôle de Recherche Interdisciplinaire sur le MédicamentLille, France
| |
Collapse
|
15
|
Munley MT, Moore JE, Walb MC, Isom SP, Olson JD, Zora JG, Kock ND, Wheeler KT, Miller MS. Cancer-prone mice expressing the Ki-rasG12C gene show increased lung carcinogenesis after CT screening exposures. Radiat Res 2011; 176:842-8. [PMID: 21962004 PMCID: PMC3244170 DOI: 10.1667/rr2649.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A >20-fold increase in X-ray computed tomography (CT) use during the last 30 years has caused considerable concern because of the potential carcinogenic risk from these CT exposures. Estimating the carcinogenic risk from high-energy, single high-dose exposures obtained from atomic bomb survivors and extrapolating these data to multiple low-energy, low-dose CT exposures using the Linear No-Threshold (LNT) model may not give an accurate assessment of actual cancer risk. Recently, the National Lung Cancer Screening Trial (NLST) reported that annual CT scans of current and former heavy smokers reduced lung cancer mortality by 20%, highlighting the need to better define the carcinogenic risk associated with these annual CT screening exposures. In this study, we used the bitransgenic CCSP-rtTA/Ki-ras mouse model that conditionally expresses the human mutant Ki-ras(G12C) gene in a doxycycline-inducible and lung-specific manner to measure the carcinogenic risk of exposure to multiple whole-body CT doses that approximate the annual NLST screening protocol. Irradiated mice expressing the Ki-ras(G12C) gene in their lungs had a significant (P = 0.01) 43% increase in the number of tumors/mouse (24.1 ± 1.9) compared to unirradiated mice (16.8 ± 1.3). Irradiated females had significantly (P < 0.005) more excess tumors than irradiated males. No tumor size difference or dose response was observed over the total dose range of 80-160 mGy for either sex. Irradiated bitransgenic mice that did not express the Ki-ras(G12C) gene had a low tumor incidence (≤ 0.1/mouse) that was not affected by exposure to CT radiation. These results suggest that (i) estimating the carcinogenic risk of multiple CT exposures from high-dose carcinogenesis data using the LNT model may be inappropriate for current and former smokers and (ii) any increased carcinogenic risk after exposure to fractionated low-dose CT-radiation may be restricted to only those individuals expressing cancer susceptibility genes in their tissues at the time of exposure.
Collapse
Affiliation(s)
- Michael T. Munley
- Department of Radiation Oncology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Joseph E. Moore
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Matthew C. Walb
- Department of Radiation Oncology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Scott P. Isom
- Department of Public Health Sciences, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - John D. Olson
- Center for Biomolecular Imaging, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - J. Gregory Zora
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Nancy D. Kock
- Department of Pathology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Kenneth T. Wheeler
- Department of Radiology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Mark Steven Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| |
Collapse
|
16
|
Kudo TA, Kanetaka H, Watanabe A, Okumoto A, Asano M, Zhang Y, Zhao F, Kano M, Shimizu Y, Tamura S, Hayashi H. Investigating bone morphogenetic protein (BMP) signaling in a newly established human cell line expressing BMP receptor type II. TOHOKU J EXP MED 2011; 222:121-9. [PMID: 20919447 DOI: 10.1620/tjem.222.121] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor β cytokine superfamily, elicit various biological effects in different tissues. BMP receptor type II (BMPRII) contains a unique carboxyl-terminal region that interacts with multiple signaling molecules. However, expression of endogenous BMPRII is low in various mammalian cell lines, which hampers the analysis of BMP signaling. Therefore, we established a human cell line expressing BMPRII tagged with a Flag epitope (BMPRII-Flag) using the tetracycline-controlled Flp-In T-REx gene expression system. The BMPRII-Flag gene was introduced into the Flp-In T-REx 293 (FT293) cell line, a derivative of human 293 embryonic kidney fibroblasts. Then we analyzed the expression of key BMP target genes, inhibitors of DNA binding (Id) family members (Id1, Id2, and Id3) and the inhibitory Smads Smad6 and Smad7, in parental FT293 cells and an established cell line, FT293-BMPRII, by quantitative real-time PCR. Tetracycline treatment significantly increased the expression of BMPRII-Flag mRNA and protein in FT293-BMPRII cells, but induced no significant changes in expression of Id1, Id2, Id3, Smad6, or Smad7 mRNA. In contrast, treatment with a BMPRII ligand BMP2 induced the expression of Id1, Id2, Id3, and Smad6 in parental FT293 cells and FT293-BMPRII cells. Tetracycline-induced BMPRII-Flag expression significantly enhanced the induction of Id1, Id3, and Smad6 mRNA expression in FT293-BMPRII cells treated with BMP2. These findings provide evidence that although BMPRII has no obvious effect on the expression of representative BMP target genes, it differentially modulates the responsiveness of target genes to BMP2.
Collapse
Affiliation(s)
- Tada-aki Kudo
- Division of Oral Physiology, Tohoku University Graduate School of Dentistry, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kher G, Trehan S, Misra A. Antisense Oligonucleotides and RNA Interference. CHALLENGES IN DELIVERY OF THERAPEUTIC GENOMICS AND PROTEOMICS 2011. [PMCID: PMC7150054 DOI: 10.1016/b978-0-12-384964-9.00007-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
18
|
Fujimoto K, Araki K, McCarthy DM, Sims JR, Ren JQ, Zhang X, Bhide PG. A transgenic mouse model of neuroepithelial cell specific inducible overexpression of dopamine D1-receptor. Neuroscience 2010; 170:961-70. [PMID: 20674683 DOI: 10.1016/j.neuroscience.2010.07.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 07/14/2010] [Accepted: 07/18/2010] [Indexed: 01/01/2023]
Abstract
Dopamine and its receptors appear in the brain during early embryonic period suggesting a role for dopamine in brain development. In fact, dopamine receptor imbalance resulting from impaired physiological balance between D1- and D2-receptor activities can perturb brain development and lead to persisting changes in brain structure and function. Dopamine receptor imbalance can be produced experimentally using pharmacological or genetic methods. Pharmacological methods tend to activate or antagonize the receptors in all cell types. In the traditional gene knockout models the receptor imbalance occurs during development and also at maturity. Therefore, assaying the effects of dopamine imbalance on specific cell types (e.g. precursor versus postmitotic cells) or at specific periods of brain development (e.g. pre- or postnatal periods) is not feasible in these models. We describe a novel transgenic mouse model based on the tetracycline dependent inducible gene expression system in which dopamine D1-receptor transgene expression is induced selectively in neuroepithelial cells of the embryonic brain at experimenter-chosen intervals of brain development. In this model, doxycycline-induced expression of the transgene causes significant overexpression of the D1-receptor and significant reductions in the incorporation of the S-phase marker bromodeoxyuridine into neuroepithelial cells of the basal and dorsal telencephalon indicating marked effects on telencephalic neurogenesis. The D1-receptor overexpression occurs at higher levels in the medial ganglionic eminence (MGE) than the lateral ganglionic eminence (LGE) or cerebral wall (CW). Moreover, although the transgene is induced selectively in the neuroepithelium, D1-receptor protein overexpression appears to persist in postmitotic cells. The mouse model can be modified for neuroepithelial cell-specific inducible expression of other transgenes or induction of the D1-receptor transgene in other cells in specific brain regions by crossbreeding the mice with transgenic mouse lines available already.
Collapse
Affiliation(s)
- K Fujimoto
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Manfredsson FP, Burger C, Rising AC, Zuobi-Hasona K, Sullivan LF, Lewin AS, Huang J, Piercefield E, Muzyczka N, Mandel RJ. Tight Long-term dynamic doxycycline responsive nigrostriatal GDNF using a single rAAV vector. Mol Ther 2009; 17:1857-67. [PMID: 19707186 DOI: 10.1038/mt.2009.196] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) gene transfer is being developed as a treatment for Parkinson's disease (PD). Due to the potential for side effects, external transgene regulation should enhance this strategy's safety profile. Here, we demonstrate dynamic control during long-term expression of GDNF using a recombinant adeno-associated virus (rAAV)-based bicistronic tetracycline (tet)-off construct. Nigrostriatal GDNF overexpression induces body weight alterations in rodents, enabling longitudinal in vivo tracking of GDNF expression after nigral vector delivery. Regulated GDNF expression was highly sensitive to dietary doxycycline (DOX), displaying undetectable striatal GDNF levels at serum DOX levels below those required for antimicrobial activity. However, in the absence of DOX, striatal GDNF levels exceeded levels required for efficacy in PD models. We also demonstrate the absence of a series of known GDNF-associated side effects when using direct intrastriatal vector delivery. Therefore, this single rAAV vector system meets most of the requirements for an experimental reagent for treatment of PD.
Collapse
Affiliation(s)
- Fredric P Manfredsson
- Department of Neuroscience, Powell Gene Therapy Center, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Mitsuhashi T, Takahashi T. Genetic regulation of proliferation/differentiation characteristics of neural progenitor cells in the developing neocortex. Brain Dev 2009; 31:553-7. [PMID: 19464833 DOI: 10.1016/j.braindev.2009.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 05/10/2009] [Indexed: 11/30/2022]
Abstract
Brain size variation among different mammals is tightly associated with different levels of cerebral function. Mechanisms that regulate the number of neurons and hence the size of the brain must be at least partially embedded within the very early phase of neocortical development, that is, embedded in proliferation/differentiation characteristics of the neural progenitor cells (NPCs) of the neocortex. Here we review a sequence of critical events through which the neocortex is formed in the embryonic forebrain, with particular emphasis on cell cycle kinetics of the NPCs that produce non-GABAergic projection neurons, the majority of neurons in the neocortex. In general, the critical parameters that determine the total number of cells produced by a given progenitor population through a sequence of cell cycles are (1) the number of cell cycles that constitute the production period and (2) the probability of cell cycle exit (Q fraction or Q) of progenitor cells for each of the cell cycles. We will also review molecular mechanisms that modulate the critical parameters above, with a special reference to the cell cycle regulatory protein p27(Kip1), inhibitor of G1 phase progression of the cell cycle. Finally the neocortical dysgenesis caused by genetic modification in mice where p27(Kip1) is either deleted or overexpressed is presented as examples of neuron number changes and resultant neocortical dysgenesis by Q fraction alteration.
Collapse
Affiliation(s)
- Takayuki Mitsuhashi
- Department of Pediatrics, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | |
Collapse
|
21
|
Dance-Barnes ST, Kock ND, Floyd HS, Moore JE, Mosley LJ, D'Agostino RB, Pettenati MJ, Miller MS. Effects of mutant human Ki-ras(G12C) gene dosage on murine lung tumorigenesis and signaling to its downstream effectors. Toxicol Appl Pharmacol 2008; 231:77-84. [PMID: 18565564 PMCID: PMC2596665 DOI: 10.1016/j.taap.2008.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 04/08/2008] [Accepted: 04/18/2008] [Indexed: 11/16/2022]
Abstract
Studies in cell culture have suggested that the level of RAS expression can influence the transformation of cells and the signaling pathways stimulated by mutant RAS expression. However, the levels of RAS expression in vivo appear to be subject to feedback regulation, limiting the total amount of RAS protein that can be expressed. We utilized a bitransgenic mouse lung tumor model that expressed the human Ki-ras(G12C) allele in a tetracycline-inducible, lung-specific manner. Treatment for 12 months with 500 microg/ml of doxycycline (DOX) allowed for maximal expression of the human Ki-ras(G12C) allele in the lung, and resulted in the development of focal hyperplasia and adenomas. We determined if different levels of mutant RAS expression would influence the phenotype of the lung lesions. Treatment with 25, 100 and 500 microg/ml of DOX resulted in dose-dependent increases in transgene expression and tumor multiplicity. Microscopic analysis of the lungs of mice treated with the 25 microg/ml dose of DOX revealed infrequent foci of hyperplasia, whereas mice treated with the 100 and 500 microg/ml doses exhibited numerous hyperplastic foci and also adenomas. Immunohistochemical and RNA analysis of the downstream effector pathways demonstrated that different levels of mutant RAS transgene expression resulted in differences in the expression and/or phosphorylation of specific signaling molecules. Our results suggest that the molecular alterations driving tumorigenesis may differ at different levels of mutant Ki-ras(G12C) expression, and this should be taken into consideration when inducible transgene systems are utilized to promote tumorigenesis in mouse models.
Collapse
Affiliation(s)
- Stephanie T Dance-Barnes
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
This overview describes issues that must be considered before attempting to express neural cDNAs in mammalian cells, including the choice of expression vector and cell type. Considerations for introducing recombinant vectors into cells are discussed along with a comparison of achieving stable or transient expression. Finally, the appropriate promoter is crucial and must be chosen to fit the design of the expression system.
Collapse
Affiliation(s)
- R L Neve
- Harvard Medical School and McLean Hospital, Belmont, Massachusetts, USA
| | | |
Collapse
|
23
|
Shockett P, Schatz D. Inducible gene expression using an autoregulatory, tetracycline-controlled system. ACTA ACUST UNITED AC 2008; Chapter 16:Unit 16.14. [PMID: 18265300 DOI: 10.1002/0471142727.mb1614s60] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The protocols in this unit describe the transfection of adherent cells and the testing of resultant clones for inducible transactivator or target gene protein expression. Stably transfected fibroblast cell lines expressing transactivator and target gene(s) can be derived by first cotransfecting pTet-tTAk and a plasmid encoding a selectable marker and obtaining stable lines with inducible transactivator expression. These lines are subsequently stably cotransfected with plasmids encoding the target gene(s) and a second selectable marker. The procedure may also be used to cotransfect pTet-tTAk with the target gene-encoding plasmid(s) and a single selectable marker plasmid. A support protocol describes methods to test stably transfected cell lines for inducible gene expression, for transient transfection and induction of tet-regulated plasmids, and for detection of the tTAk gene in cells (or transgenic mice).
Collapse
Affiliation(s)
- Penny Shockett
- Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
24
|
Shockett P, Schatz D. Inducible gene expression using an autoregulatory, tetracycline-controlled system. CURRENT PROTOCOLS IN CELL BIOLOGY 2008; Chapter 20:20.8.1-20.8.10. [PMID: 18228465 DOI: 10.1002/0471143030.cb2008s27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The protocols in this unit describe the transfection of adherent cells and the testing of resultant clones for inducible transactivator or target gene protein expression. Stably transfected fibroblast cell lines expressing transactivator and target gene(s) can be derived by first cotransfecting pTet-tTAk and a plasmid encoding a selectable marker and obtaining stable lines with inducible transactivator expression. These lines are subsequently stably cotransfected with plasmids encoding the target gene(s) and a second selectable marker. The procedure may also be used to cotransfect pTet-tTAk with the target gene-encoding plasmid(s) and a single selectable marker plasmid. A support protocol describes methods to test stably transfected cell lines for inducible gene expression, for transient transfection and induction of tet-regulated plasmids, and for detection of the tTAk gene in cells (or transgenic mice).
Collapse
Affiliation(s)
| | - David Schatz
- Howard Hughes Medical Institute and Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
25
|
Winkeler A, Sena-Esteves M, Paulis LE, Li H, Waerzeggers Y, Rückriem B, Himmelreich U, Klein M, Monfared P, Rueger MA, Heneka M, Vollmar S, Hoehn M, Fraefel C, Graf R, Wienhard K, Heiss WD, Jacobs AH. Switching on the lights for gene therapy. PLoS One 2007; 2:e528. [PMID: 17565381 PMCID: PMC1885827 DOI: 10.1371/journal.pone.0000528] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 04/30/2007] [Indexed: 11/19/2022] Open
Abstract
Strategies for non-invasive and quantitative imaging of gene expression in vivo have been developed over the past decade. Non-invasive assessment of the dynamics of gene regulation is of interest for the detection of endogenous disease-specific biological alterations (e.g., signal transduction) and for monitoring the induction and regulation of therapeutic genes (e.g., gene therapy). To demonstrate that non-invasive imaging of regulated expression of any type of gene after in vivo transduction by versatile vectors is feasible, we generated regulatable herpes simplex virus type 1 (HSV-1) amplicon vectors carrying hormone (mifepristone) or antibiotic (tetracycline) regulated promoters driving the proportional co-expression of two marker genes. Regulated gene expression was monitored by fluorescence microscopy in culture and by positron emission tomography (PET) or bioluminescence (BLI) in vivo. The induction levels evaluated in glioma models varied depending on the dose of inductor. With fluorescence microscopy and BLI being the tools for assessing gene expression in culture and animal models, and with PET being the technology for possible application in humans, the generated vectors may serve to non-invasively monitor the dynamics of any gene of interest which is proportionally co-expressed with the respective imaging marker gene in research applications aiming towards translation into clinical application.
Collapse
Affiliation(s)
- Alexandra Winkeler
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Miguel Sena-Esteves
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Leonie E.M. Paulis
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Hongfeng Li
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Yannic Waerzeggers
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Benedikt Rückriem
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Uwe Himmelreich
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Markus Klein
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Parisa Monfared
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Maria A. Rueger
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Michael Heneka
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Stefan Vollmar
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Mathias Hoehn
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Cornel Fraefel
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Rudolf Graf
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Klaus Wienhard
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Wolf D. Heiss
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Andreas H. Jacobs
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
26
|
Stringent and reproducible tetracycline-regulated transgene expression by site-specific insertion at chromosomal loci with pre-characterised induction characteristics. BMC Mol Biol 2007; 8:30. [PMID: 17493262 PMCID: PMC1884169 DOI: 10.1186/1471-2199-8-30] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 05/10/2007] [Indexed: 12/16/2022] Open
Abstract
Background The ability to regulate transgene expression has many applications, mostly concerning the analysis of gene function. Desirable induction characteristics, such as low un-induced expression, high induced expression and limited cellular heterogeneity, can be seriously impaired by chromosomal position effects at the site of transgene integration. Many clones may therefore need to be screened before one with optimal induction characteristics is identified. Furthermore, such screens must be repeated for each new transgene investigated, and comparisons between clones with different transgenes is complicated by their different integration sites. Results To circumvent these problems we have developed a "screen and insert" strategy in which clones carrying a transgene for a fluorescent reporter are first screened for those with optimal induction characteristics. Site-specific recombination (SSR) is then be used repeatedly to insert any new transgene at the reporter transgene locus of such clones so that optimal induction characteristics are conferred upon it. Here we have tested in a human fibrosarcoma cell line (HT1080) two of many possible implementations of this approach. Clones (e.g. Rht14-10) in which a GFP reporter gene is very stringently regulated by the tetracycline (tet) transactivator (tTA) protein were first identified flow-cytometrically. Transgenes encoding luciferase, I-SceI endonuclease or Rad52 were then inserted by SSR at a LoxP site adjacent to the GFP gene resulting stringent tet-regulated transgene expression. In clone Rht14-10, increases in expression from essentially background levels (+tet) to more than 104-fold above background (-tet) were reproducibly detected after Cre-mediated insertion of either the luciferase or the I-SceI transgenes. Conclusion Although previous methods have made use of SSR to integrate transgenes at defined sites, none has effectively combined this with a pre-selection step to identify integration sites that support optimal regulatory characteristics. Rht14-10 and similar HT1080-derived clones can now be used in conjunction with a convenient delivery vector (pIN2-neoMCS), in a simple 3-step protocol leading to stringent and reproducible transgene regulation. This approach will be particularly useful for transgenes whose products are very active at low concentrations and/or for comparisons of multiple related transgenes.
Collapse
|
27
|
Welman A, Barraclough J, Dive C. Tetracycline regulated systems in functional oncogenomics. TRANSLATIONAL ONCOGENOMICS 2007; 2:17-33. [PMID: 23645981 PMCID: PMC3634622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The increasing number of proteomic and DNA-microarray studies is continually providing a steady acquisition of data on the molecular abnormalities associated with human tumors. Rapid translation of this accumulating biological information into better diagnostics and more effective cancer therapeutics in the clinic depends on the use of robust function-testing strategies. Such strategies should allow identification of molecular lesions that are essential for the maintenance of the transformed phenotype and enable validation of potential drug-targets. The tetracycline regulated gene expression/ suppression systems (Tet-systems) developed and optimized by bioengineers over recent years seem to be very well suited for the function-testing purposes in cancer research. We review the history and latest improvements in Tet-technology in the context of functional oncogenomics.
Collapse
Affiliation(s)
- Arkadiusz Welman
- Cancer Research U.K., Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom.,Correspondence: Arkadiusz Welman, Ph.D, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom. Tel: +44 161 446 8104; Fax: +44 161 446 3109;
| | - Jane Barraclough
- Cancer Research U.K., Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom
| | - Caroline Dive
- Cancer Research U.K., Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, United Kingdom
| |
Collapse
|
28
|
Shen Q, Fan L, Newburger PE. Nuclease sensitive element binding protein 1 associates with the selenocysteine insertion sequence and functions in mammalian selenoprotein translation. J Cell Physiol 2006; 207:775-83. [PMID: 16508950 PMCID: PMC3730826 DOI: 10.1002/jcp.20619] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Biosynthesis of selenium-containing proteins requires insertion of the unusual amino acid selenocysteine by alternative translation of a UGA codon, which ordinarily serves as a stop codon. In eukaryotes, selenoprotein translation depends upon one or more selenocysteine insertion sequence (SECIS) elements located in the 3'-untranslated region of the mRNA, as well as several SECIS-binding proteins. Our laboratory has previously identified nuclease sensitive element binding protein 1 (NSEP1) as another SECIS-binding protein, but evidence has been presented both for and against its role in SECIS binding in vivo and in selenoprotein translation. Our current studies sought to resolve this controversy, first by investigating whether NSEP1 interacts closely with SECIS elements within intact cells. After reversible in vivo cross-linking and ribonucleoprotein immunoprecipitation, mRNAs encoding two glutathione peroxidase family members co-precipitated with NSEP1 in both human and rat cell lines. Co-immunoprecipitation of an epitope-tagged GPX1 construct depended upon an intact SECIS element in its 3'-untranslated region. To test the functional importance of this interaction on selenoprotein translation, we used small inhibitory RNAs to reduce the NSEP1 content of tissue culture cells and then examined the effect of that reduction on the activity of a SECIS-dependent luciferase reporter gene for which expression depends upon readthrough of a UGA codon. Co-transfection of small inhibitory RNAs directed against NSEP1 decreased its expression by approximately 50% and significantly reduced luciferase activity. These studies demonstrate that NSEP1 is an authentic SECIS binding protein that is structurally associated with the selenoprotein translation complex and functionally involved in the translation of selenoproteins in mammalian cells.
Collapse
Affiliation(s)
- Qichang Shen
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Lin Fan
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Peter E. Newburger
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
- Correspondence to: Dr. Peter E. Newburger, Department of Pediatrics, LRB 404, 364 Plantation Street, Worcester, Massachusetts 01605.
| |
Collapse
|
29
|
Hale MA, Kagami H, Shi L, Holland AM, Elsässer HP, Hammer RE, MacDonald RJ. The homeodomain protein PDX1 is required at mid-pancreatic development for the formation of the exocrine pancreas. Dev Biol 2005; 286:225-37. [PMID: 16126192 DOI: 10.1016/j.ydbio.2005.07.026] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 06/29/2005] [Accepted: 07/01/2005] [Indexed: 10/25/2022]
Abstract
The homeoprotein PDX1 is expressed throughout pancreatic development and is thought to play important roles at multiple stages. We describe the properties of a tet-off regulatory scheme to manage the expression of Pdx1 in utero. Cessation of Pdx1 expression at increasingly later gestational times blocked pancreatic development at progressive and morphologically distinct stages and provided the opportunity to assess the requirement for Pdx1 at each stage. Embryonic PDX1 is depleted below effective levels within 1 day of the initiation of doxycycline treatment of pregnant mice. We show that PDX1, which is necessary for early pancreatic development, is also required later for the genesis of acinar tissue, the compartment of the pancreas that produces digestive enzymes. Without PDX1, acini do not form; the precursor epithelium continues to grow and branch, creating a truncated ductal tree comprising immature duct-like cells. The bHLH factor PTF1a, a critical regulator of acinar development, is not expressed and cells producing digestive enzymes are rare. This approach should be generally applicable to study the in vivo functions of other developmental regulators with multiple, temporally distinct roles.
Collapse
Affiliation(s)
- Michael A Hale
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9148, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Zhang QH, Wang LL, Cao L, Peng C, Li XL, Tang K, Li WF, Liao P, Wang JR, Li GY. Study of a novel brain relatively specific gene LRRC4 involved in glioma tumorigenesis suppression using the Tet-on system. Acta Biochim Biophys Sin (Shanghai) 2005; 37:532-40. [PMID: 16077900 DOI: 10.1111/j.1745-7270.2005.00079.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
LRRC4 is a novel relatively specific gene, which displays significant down-regulation in primary brain tumor biopsies and has the potential to suppress brain tumor growth. In this study, we investigated the growth inhibitory effect of LRRC4 on tumorigencity in vivo and on cell proliferation in vitro by a tetracycline-inducible expression system. Results showed that LRRC4 significantly reduced the growth and malignant grade of xenografts arising from glioblastoma U251MG cells. Cell proliferation was markedly inhibited after U251MG Tet-on-LRRC4 cell induction with doxycycline. Flow cytometry and Western blot analysis demonstrated that LRRC4 mediated a delay of the cell cycle in late G1, possibly through up-regulating the expressions of p21Waf1/cip1 and p27Kip1 and down-regulating the expressions of cyclin-dependent kinase 2, retinoblastoma protein and epidermal growth factor receptors. Together, these findings provide clues to the function of LRRC4 as a negative regulator of cell growth and underscore a link between the above-mentioned cyclins, cyclin-associated molecules and tumorigenicity.
Collapse
Affiliation(s)
- Qiu-Hong Zhang
- Cancer Research Institute, Central South University, Changsha 410078, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhang Q, Wang J, Fan S, Wang L, Cao L, Tang K, Peng C, Li Z, Li W, Gan K, Liu Z, Li X, Shen S, Li G. Expression and functional characterization ofLRRC4, a novel brain-specific member of the LRR superfamily. FEBS Lett 2005; 579:3674-82. [PMID: 15967442 DOI: 10.1016/j.febslet.2005.05.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 04/20/2005] [Accepted: 05/09/2005] [Indexed: 01/08/2023]
Abstract
LRRC4, a novel member of LRR superfamily thought to be involved in development and tumorigenesis of the nervous tissue, has the potential to suppress tumorigenesis and cell proliferation of U251MG cells. This study aimed at revealing the correlation between expression of LRRC4 and the maintenance of normal function and tumorigenesis suppression within the central nervous system. We systematically analyzed the expression and tissue distributions of the gene in tissues. Results showed that LRRC4 expression was limited to normal adult brain, both in human and in mouse, and exhibited a development-regulated pattern, but was down-regulated in brain tumor tissues and U251MG cell line. Furthermore, dynamic alterations in gene expression associated with cell cycle progression were investigated by using Tet-on system. Results showed that LRRC4 induced a cell cycle delay at the late G1 phase, probably through the alteration of the expression of different cell cycle regulating proteins responsible for mediating G1-S progression, such as p21(Waf1/Cip1) and p27(Kip1), Cdk2 and PCNA, p-ERK1/2. These findings suggest that LRRC4 may play an important role in maintaining normal function and suppressing tumorigenesis in the central nervous system.
Collapse
Affiliation(s)
- Qiuhong Zhang
- Cancer Research Institute, Central South University, Changsha 410078, Hunan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Godambe SA, Knapp KM, Meals EA, English BK. Role of vav1 in the lipopolysaccharide-mediated upregulation of inducible nitric oxide synthase production and nuclear factor for interleukin-6 expression activity in murine macrophages. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 11:525-31. [PMID: 15138177 PMCID: PMC404562 DOI: 10.1128/cdli.11.3.525-531.2004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
vav1 has been shown to play a key role in lymphocyte development and activation, but its potential importance in macrophage activation has received little attention. We have previously reported that exposure of macrophages to bacterial lipopolysaccharide (LPS) leads to increased activity of hck and other src-related tyrosine kinases and to the prompt phosphorylation of vav1 on tyrosine. In this study, we tested the role of vav1 in macrophage responses to LPS, focusing on the upregulation of nuclear factor for interleukin-6 expression (NF-IL-6) activity and inducible nitric oxide synthase (iNOS) protein accumulation in RAW-TT10 murine macrophages. We established a series of stable cell lines expressing three mutant forms of vav1 in a tetracycline-regulatable fashion: (i) a form producing a truncated protein, vavC; (ii) a form containing a point mutation in the regulatory tyrosine residue, vavYF174; and (iii) a form with an in-frame deletion of 6 amino acids required for the guanidine nucleotide exchange factor (GEF) activity of vav1 for rac family GTPases, vavGEFmt. Expression of the truncated mutant (but not the other two mutants) has been reported to interfere with T-cell activation. In contrast, we now demonstrate that expression of any of the three mutant forms of vav1 in RAW-TT10 cells consistently inhibited LPS-mediated increases in iNOS protein accumulation and NF-IL-6 activity. These data provide direct evidence for a role for vav1 in LPS-mediated macrophage activation and iNOS production and suggest that vav1 functions in part via activation of NF-IL-6. Furthermore, these findings indicate that the GEF activity of vav1 is required for its ability to mediate macrophage activation by LPS.
Collapse
Affiliation(s)
- Sandip A Godambe
- Department of Pediatrics, Children's Foundation Research Center, Le Bonheur Children's Medical Center, 50 N. Dunlap, Memphis, TN 38103, USA
| | | | | | | |
Collapse
|
33
|
Peng H, Usas A, Gearhart B, Young B, Olshanski A, Huard J. Development of a self-inactivating tet-on retroviral vector expressing bone morphogenetic protein 4 to achieve regulated bone formation. Mol Ther 2005; 9:885-94. [PMID: 15194055 DOI: 10.1016/j.ymthe.2004.02.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2003] [Accepted: 02/20/2004] [Indexed: 11/19/2022] Open
Abstract
The aims of this study were to explore the possibility of improving the design of self-inactivating (SI) retroviral vectors and to develop an SI vector that would allow optimal tet-on-regulated therapeutic gene expression. To minimize any interference between the viral promoter and the inducible promoter, we deleted different regulatory elements in the 3'LTR and examined their effects on transgene expression in transfected or transduced cells. In transfected cells, such deletions reduced the transgene expression. The insertion of a polyadenylation sequence could not completely compensate for this effect. We observed three patterns of transgene expression in cells transduced with these tet-on retroviral vectors: (1) high levels of both basal and inducible expression, (2) low levels of both basal and inducible expression, and (3) low levels of basal and high levels of inducible expression. After using the optimal vector to transduce muscle-derived stem cells, we were able to regulate the strong in vitro expression of transgenes-including enhanced green fluorescent protein and bone morphogenetic protein 4-via the addition or withdrawal of doxycycline (Dox). Implantation of the transduced cells and subsequent Dox-dependent induction of gene expression resulted in bone formation in vivo. Thus, we have developed an optimal SI retroviral vector that maintains a high titer, efficiently transduces muscle-derived stem cells, and enables both high levels of inducible gene expression in vitro and robust regulated bone formation in vivo.
Collapse
Affiliation(s)
- Hairong Peng
- Growth and Development Laboratory, Children's Hospital of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
34
|
Takács K, Du Roure C, Nabarro S, Dillon N, McVey JH, Webster Z, Macneil A, Bartók I, Higgins C, Gray D, Merkenschlager M, Fisher AG. The regulated long-term delivery of therapeutic proteins by using antigen-specific B lymphocytes. Proc Natl Acad Sci U S A 2004; 101:16298-303. [PMID: 15520381 PMCID: PMC528951 DOI: 10.1073/pnas.0405271101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Memory lymphocytes are important mediators of the immune response. These cells are long-lived and undergo clonal expansion upon reexposure to specific antigen, differentiating into effector cells that secrete Ig or cytokines while maintaining a residual pool of memory T and B lymphocytes. Here, the ability of antigen-specific lymphocytes to undergo repeated cycles of antigen-driven clonal expansion and contraction is exploited in a therapeutic protocol aimed at regulating protein delivery. The principle of this strategy is to introduce genes encoding proteins of therapeutic interest into a small number of antigen-specific B lymphocytes. Output of therapeutic protein can then be regulated in vivo by manipulating the size of the responder population by antigen challenge. To evaluate whether such an approach is feasible, we developed a mouse model system in which Emu- and Iglambda-based vectors were used to express human erythropoietin (hEPO) gene in B lymphocytes. These mice were then immunized with the model antigen phycoerythrin (PE), and immune splenocytes (or purified PE-specific B lymphocytes) were adoptively transferred to normal or mutant (EPO-deficient) hosts. High levels of hEPO were detected in the serum of adoptively transferred normal mice after PE administration, and this responsiveness was maintained for several months. Similarly, in EPO-deficient anemic recipients, antigen-driven hEPO expression was shown to restore hematocrit levels to normal. These results show that antigen-mediated regulation of memory lymphocytes can be used as a strategy for delivering therapeutic proteins in vivo.
Collapse
Affiliation(s)
- Katalin Takács
- Lymphocyte Development Group, Gene Regulation and Chromatin Group, Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College of Science, Technology, Du Cane Road, London W12 ONN, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kanwal C, Mu S, Kern SE, Lim CS. Bidirectional on/off switch for controlled targeting of proteins to subcellular compartments. J Control Release 2004; 98:379-93. [PMID: 15312994 DOI: 10.1016/j.jconrel.2004.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2004] [Accepted: 05/20/2004] [Indexed: 01/21/2023]
Abstract
A regulatable fusion protein was constructed for controlling the localization of plasmid products. A ligand-inducible nuclear localization signal, nuclear export signal (NES) and a truncated form of the ligand binding domain of the progesterone receptor were attached to the desired protein. Enhanced green fluorescent protein (EGFP) was used as a model protein and its trafficking between the nucleus and cytoplasm was studied using fluorescence microscopy in response to the ligand, mifepristone. It was found that the protein trafficking into the nucleus was dose dependent with ligand concentration. Increasing the ligand dose from 1 to 100 nM enhanced import and reduced the rate of export of the fusion protein from the nucleus to the cytoplasm. This study demonstrates the feasibility of using an export signal and a ligand-inducible nuclear import signal as a bi-directional on/off switch with potential use for controlled targeting of therapeutic proteins to subcellular compartments.
Collapse
Affiliation(s)
- C Kanwal
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 421 Wakara Way #318, Salt Lake City, UT 84108, USA
| | | | | | | |
Collapse
|
36
|
Jääskeläinen I, Lappalainen K, Honkakoski P, Urtti A. Requirements for delivery of active antisense oligonucleotides into cells with lipid carriers. Methods Enzymol 2004; 387:210-30. [PMID: 15172166 DOI: 10.1016/s0076-6879(04)87013-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
|
37
|
Abstract
The lung is a complex organ consisting of numerous cell types that function to ensure sufficient gas exchange to oxygenate the blood. In order to accomplish this function, the lung must be exposed to the external environment and at the same time maintain a homeostatic balance between its function in gas exchange and the maintenance of inflammatory balance. During the past two decades, as molecular methodologies have evolved with the sequencing of entire genomes, the use of in vivo models to elucidate the molecular mechanisms involved in pulmonary physiology and disease have increased. The mouse has emerged as a potent model to investigate pulmonary physiology due to the explosion in molecular methods that now allow for the developmental and tissue-specific regulation of gene transcription. Initial efforts to manipulate gene expression in the mouse genome resulted in the generation of transgenic mice characterized by the constitutive expression of a specific gene and knockout mice characterized by the ablation of a specific gene. The utility of these original mouse models was limited, in many cases, by phenotypes resulting in embryonic or neonatal lethality that prevented analysis of the impact of the genetic manipulation on pulmonary biology. Second-generation transgenic mouse models employ multiple strategies that can either activate or silence gene expression thereby providing extensive temporal and spatial control of the experimental parameters of gene expression. These highly regulated mouse models are intended to serve as a foundation for further investigation of the molecular basis of human disease such as tumorigenesis. This review describes the principles, progress, and application of systems that are currently employed in the conditional regulation of gene expression in the investigation of lung cancer.
Collapse
Affiliation(s)
- I Kwak
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
38
|
Shockett PE, Zhou S, Hong X, Schatz DG. Partial reconstitution of V(D)J rearrangement and lymphocyte development in RAG-deficient mice expressing inducible, tetracycline-regulated RAG transgenes. Mol Immunol 2004; 40:813-29. [PMID: 14687938 DOI: 10.1016/j.molimm.2003.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Previously, we described a tetracycline-based autoregulatory system for inducible gene expression in mammalian cells and transgenic mice [Proc. Natl. Acad. Sci. U.S.A. 92 (1995) 6522]. We have tested the ability of this system to drive functional expression in vivo of the V(D)J recombination activating genes, RAG1 and RAG2. In induced transgenic mice, transgenic RAG1 and RAG2 mRNA is observed in thymus and spleen, and expression of both transgenes on the RAG1 or RAG2 knockout backgrounds allows partial, inducible, lymphocyte reconstitution. In thymus and peripheral lymphoid organs of reconstituted animals, cells expressing CD4 and/or CD8 on their surface, also express CD3 and TCR beta chain. In these animals, V(D)J rearrangements are detected in thymus, lymph nodes, and spleen at the TRB locus, and in thymus and lymph nodes at the TRD locus. At the TRA locus, broken ends at V(D)J recombination signals are detected only in thymus, as are reciprocal signal joint products derived from deletional rearrangement. T cell reconstitution occurs in these animals whether they are induced in utero during development, or shortly after birth. A low level of B cell reconstitution is also observed. B220+IgM+ cells are observed in spleen only in induced animals, and rearrangements at IGH and IGK loci are detected in bone marrow and spleen. Broken signal ends at the IGK locus, are not detected in peripheral lymphoid organs. Inducible reconstitution of normal levels of serum immunoglobulin, including heavy chain class switch isotype variants is also observed in these animals. Further, these transgenes do not appear to interfere with lymphocyte development mediated by functionally rearranged TRB chain or IGH chain transgenes in RAG-deficient animals. These mice provide a unique system for the inducible activation of V(D)J recombination and the development of primary lymphocytes.
Collapse
Affiliation(s)
- Penny E Shockett
- Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | |
Collapse
|
39
|
Berens C, Hillen W. Gene regulation by tetracyclines. Constraints of resistance regulation in bacteria shape TetR for application in eukaryotes. EUROPEAN JOURNAL OF BIOCHEMISTRY 2003; 270:3109-21. [PMID: 12869186 DOI: 10.1046/j.1432-1033.2003.03694.x] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The Tet repressor protein (TetR) regulates transcription of a family of tetracycline (tc) resistance determinants in Gram-negative bacteria. The resistance protein TetA, a membrane-spanning H+-[tc.M]+ antiporter, must be sensitively regulated because its expression is harmful in the absence of tc, yet it has to be expressed before the drugs' concentration reaches cytoplasmic levels inhibitory for protein synthesis. Consequently, TetR shows highly specific tetO binding to reduce basal expression and high affinity to tc to ensure sensitive induction. Tc can cross biological membranes by diffusion enabling this inducer to penetrate the majority of cells. These regulatory and pharmacological properties are the basis for application of TetR to selectively control the expression of single genes in lower and higher eukaryotes. TetR can be used for that purpose in some organisms without further modifications. In mammals and in a large variety of other organisms, however, eukaryotic transcriptional activator or repressor domains are fused to TetR to turn it into an efficient regulator. Mechanistic understanding and the ability to engineer and screen for mutants with specific properties allow tailoring of the DNA recognition specificity, the response to inducer tc and the dimerization specificity of TetR-based eukaryotic regulators. This review provides an overview of the TetR properties as they evolved in bacteria, the functional modifications necessary to transform it into a convenient, specific and efficient regulator for use in eukaryotes and how the interplay between structure--function studies in bacteria and specific requirements of particular applications in eukaryotes have made it a versatile and highly adaptable regulatory system.
Collapse
Affiliation(s)
- Christian Berens
- Lehrstuhl für Mikrobiologie, Institut für Mikrobiologie, Biochemie und Genetik, Friedrich-Alexander Universität Erlangen-Nürnberg, Germany
| | | |
Collapse
|
40
|
Tietge UJF, Kozarsky KF, Donahee MH, Rader DJ. A tetracycline-regulated adenoviral expression system for in vivo delivery of transgenes to lung and liver. J Gene Med 2003; 5:567-75. [PMID: 12825196 DOI: 10.1002/jgm.384] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Recombinant adenoviruses are an established tool for somatic gene transfer to multiple cell types in animals as well as in tissue culture. However, generation of adenoviruses expressing transgenes that are potentially toxic to the host cell line represents a practical problem. The aim of this study was to construct an adenoviral expression system that prevents transgene expression during the generation and propagation of the virus, and allows efficient gene transfer to lung and liver, major target organs of gene therapy. METHODS Using the tet-off system we constructed tetracycline (tet) regulatable recombinant adenoviruses expressing the marker gene LacZ (Adtet-off.LacZ) as well as a secretory protein, human group IIA secretory phospholipase A(2) (Adtet-off.hsPLA(2)). Expression (Western blot, activity assay) was tested in vitro (HeLa cells), and in vivo by gene transfer to lung and liver. RESULTS Without addition of tetracycline we demonstrated expression of LacZ (Adtet-off.LacZ) and sPLA(2) (Adtet-off.hsPLA(2)) in HeLa cells. Providing additional tet-transactivator (tTA) protein either by stable transfection or coinfection with a tTA-expressing adenovirus resulted in a further increase of LacZ and sPLA(2) expression. Transgene expression in vitro was eliminated by the addition of tetracycline to the culture medium. Adtet-off.LacZ and Adtet-off.hsPLA(2) allowed successful gene transfer in vivo to lung and liver. While the expression was highly efficient within the lungs, however, additional tTA was necessary to achieve high-level expression within liver. CONCLUSIONS Tet-regulatable adenoviral expression systems may facilitate the construction of recombinant adenoviruses encoding potentially toxic transgenes and permit regulated transgene expression.
Collapse
Affiliation(s)
- Uwe J F Tietge
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6160, USA
| | | | | | | |
Collapse
|
41
|
Lin W, Albanese C, Pestell RG, Lawrence DS. Spatially discrete, light-driven protein expression. CHEMISTRY & BIOLOGY 2002; 9:1347-53. [PMID: 12498888 DOI: 10.1016/s1074-5521(02)00288-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Transgene-based inducible expression systems offer the potential to study the influence of any gene at any point during an organism's lifetime. However, the expression of individual genes is both temporally and spatially (i.e., cell/tissue)-regulated. The inducible gene expression systems devised to date do not offer fine spatial control over gene expression. We describe herein the creation and study of a light-activatable, ecdysone-inducible gene expression system. We have constructed the first example of a caged ecdysteroid, which is virtually inactive as an inducing agent in a luciferase-based gene expression system. However, upon exposure to brief illumination, the caged ecdysteroid is rapidly converted into active beta-ecdysone. Caged beta-ecdysone is cell permeable, can be intracellularly photouncaged, and, in combination with spot illumination, can be used to drive spatially discrete protein expression in a multicellular setting.
Collapse
Affiliation(s)
- Weiying Lin
- Department of Biochemistry, The Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
42
|
Liu J, Johnston MR. Animal models for studying lung cancer and evaluating novel intervention strategies. Surg Oncol 2002; 11:217-27. [PMID: 12450558 DOI: 10.1016/s0960-7404(02)00053-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The pathogenesis of lung cancer progression, invasion and metastasis remains undefined. Clinically relevant laboratory models of the disease could greatly facilitate its clarification. Model systems of lung cancer that accurately reflect different biologic properties and disease stages are necessary to ensure proper experimental design of studies aimed at increasing our understanding of the disease. Such models are also essential tools to accelerate development of new therapies for lung cancer. In this review we summarize the available lung cancer model systems in use today and define both their utility and limitations.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Thoracic Surgery, Toronto General Hospital, Division of Surgical Oncology, Princess Margaret Hospital, University of Toronto, Ont., Canada
| | | |
Collapse
|
43
|
Teng PI, Dichiara MR, Kömüves LG, Abe K, Quertermous T, Topper JN. Inducible and selective transgene expression in murine vascular endothelium. Physiol Genomics 2002; 11:99-107. [PMID: 12388791 DOI: 10.1152/physiolgenomics.00059.2002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have developed a system utilizing the murine Tie2 promoter/enhancer coupled with the "tetracycline-on" regulatory elements to create a model that allows regulated and selective expression of a beta-galactosidase (betaGal) reporter transgene in the adult murine vascular endothelium. Two independent lines of viable and fertile mice were characterized, and they exhibit minimal betaGal expression under basal conditions. In response to exogenous doxycycline (Dox), selective expression of betaGal was demonstrated in the vascular endothelium of all tissues examined. En face analyses of the aorta and its principle branches indicate that the vast majority of lumenal endothelial cells express the transgene. Inducible betaGal expression also extends to the endocardium and the microvasculature of all organs. There is no evidence of specific transgene expression in nonendothelial cell types. Induction of the betaGal was effectively achieved after 3 days of oral Dox treatment and persisted for over 3 mo with continuous administration. This model can now be widely applied to study the role of specific genes in the phenotype of adult murine vasculature.
Collapse
Affiliation(s)
- Peter I Teng
- Millennium Pharmaceuticals, South San Francisco 94080, USA
| | | | | | | | | | | |
Collapse
|
44
|
Approaches to Modeling Stromal-Epithelial Interactions. J Urol 2002. [DOI: 10.1097/00005392-200209000-00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Abstract
PURPOSE Techniques that can be used to examine the molecular mechanisms of stromal-epithelial interactions are described. MATERIALS AND METHODS A historical perspective of available techniques is provided. Recent developments and examples are used to illustrate and provide descriptive literature references for these methods. Since the possibilities for manipulating experimental systems are enormous and rapidly expanding, the reader should be aware that this review is an overview of how data have been and could be obtained rather than a comprehensive listing of what has been achieved. This review focuses on studies performed in the organs of the urogenital tract to illustrate techniques that are available.RESULTS Recent technological innovations have impacted our ability to manipulate specific components and pathways of stromal-epithelial interactions. They include rapid developments in transgenic and gene knockout mouse technology, and the development of highly efficacious gene delivery and expression systems. CONCLUSIONS These technologies have the potential to transform our understanding of the mechanistic basis of intercellular communication and point the way toward new therapeutic approaches for benign and malignant proliferative conditions.
Collapse
Affiliation(s)
- Simon W Hayward
- Department of Urologic Surgery, Valderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Sommer B, Rinsch C, Payen E, Dalle B, Schneider B, Déglon N, Henri A, Beuzard Y, Aebischer P. Long-term doxycycline-regulated secretion of erythropoietin by encapsulated myoblasts. Mol Ther 2002; 6:155-61. [PMID: 12161181 DOI: 10.1006/mthe.2002.0646] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We developed an ex vivo gene therapy approach for the regulated delivery of therapeutic proteins based on the implantation of encapsulated, genetically engineered C(2)C(12) myoblasts. We investigated doxycycline-based regulation of gene expression to modulate the secretion of erythropoietin (EPO) from encapsulated myoblasts in a mouse model. An autoregulatory tet-off system provided high induction levels with low basal expression in the noninduced state. Stable C(2)C(12) clones constitutively secreted between 25 and 50 IU mouse EPO/10(6)cells/24 hours in the on-state. The clone C15, selected for its in vivo survival characteristics, displayed a desirable secretion profile when encapsulated. Devices released 5 IU EPO per capsule in the on-state, with EPO levels being undetectable upon the addition of doxycycline (dox). Capsules subcutaneously implanted in DBA/2J mice demonstrated a tightly regulated secretion of EPO through up to four on-off cycles during a period lasting 40 weeks. Hematocrits could be modulated between basal levels (40-50%) and elevated levels (70-90%) through the presence or absence of dox in the drinking water. Hematocrit returned to normal levels, paralleling the kinetics observed following capsule explantation, 6 to 8 weeks following dox administration to polycythemic mice. The results of this study suggest that encapsulation and implantation of a tet-off regulated C(2)C(12) cell clone represents a safe method for the controlled long-term delivery of proteins in vivo.
Collapse
Affiliation(s)
- Birgit Sommer
- Division of Surgical Research and Gene Therapy Center, Lausanne Medical School, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Grimberg A, Liu B, Bannerman P, El-Deiry WS, Cohen P. IGFBP-3 mediates p53-induced apoptosis during serum starvation. Int J Oncol 2002; 21:327-35. [PMID: 12118329 PMCID: PMC4152903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Insulin-like growth factor binding protein (IGFBP)-3, a p53-response gene, can induce apoptosis in an IGF-independent manner. Here we demonstrate that IGFBP-3 mediates p53-induced apoptosis during serum starvation using two foil neoplastic cell models: one which introduces p53 activity and one which eliminates it. We created a doxycycline-inducible p53 model from the p53-negative PC-3 prostate cancer cell line. Doxycycline treatment increased both p53 and IGFBP-3 levels. It also augmented apoptosis, but not during insulin-like growth factor-I co-treatment. In a second model, lung carcinoma H460 cells expressing fully functional p53 were stably transfected with E6, which targets p53 for degradation. H460-E6 cells contained less p53 and IGFBP-3 than control neo-transfected cells, and proteasome blockade restored both. In serum deprivation, H460-E6 cells had enhanced growth and less apoptosis than did H460-neo cells. Reductions in H460-neo apoptosis, comparable in magnitude to H460-E6, were achieved by adding anti-IGFBP-3-antibody or IGFBP-3 antisense oligomers, but not non-specific immunoglobulin or IGFBP-3 sense oligomers. In summary, turning p53 in two foil neoplastic cell models induced IGFBP-3 expression and increased apoptosis during serum starvation, an effect inhibited by insulin-like growth factor-I treatment and specific IGFBP-3 blockade. This is the first demonstration of inhibition of p53 action by antagonizing IGFBP-3.
Collapse
Affiliation(s)
- Adda Grimberg
- Division of Pediatric Endocrinology, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
48
|
Shi Y, Koh JT. Functionally orthogonal ligand-receptor pairs for the selective regulation of gene expression generated by manipulation of charged residues at the ligand-receptor interface of ER alpha and ER beta. J Am Chem Soc 2002; 124:6921-8. [PMID: 12059215 DOI: 10.1021/ja016897x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The reengineering of protein-small molecule interfaces represents a powerful tool of chemical biology. For many applications it is necessary to engineer receptors so that they do not interact with their endogenous ligands but are highly responsive to designed ligand analogues, which in turn do not interact with endogenous proteins. The chemical design strategy used to reengineer protein-small molecule interfaces is particularly challenging for interfaces involving relatively plastic receptor binding sites and therefore presents a unique challenge in molecular design. In this study we explore the scope and limitations of a new strategy for manipulating polar/charged residues across the ligand receptor interface of estradiol (E2) and the estrogen receptor (ER). Carboxylate-functionalized E2 analogues can activate ER alpha(Glu353-->Ala) and ER beta(Glu305-->Ala) with very large selectivites, demonstrating that this design strategy is extendable to other members of the steroid hormone receptor family. Neutral E2 analogues were found to complement ER alpha(E353A) with similar potencies but with generally lower selectivities. This suggests that the high selectivity observed with ligand-receptor pairs generated by exchanging charged residues across ligand-receptor interfaces is only due in part to their complementary shapes and that appropriate introduction of charged functionality on the ligand can provide substantial enhancement of selectivity by decreasing the engineered ligands affinity for the endogenous receptor. Attempts to modify the cationic residues by complementing Arg394-->Ala or Arg394-->Glu were not successful.
Collapse
Affiliation(s)
- Youheng Shi
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | | |
Collapse
|
49
|
Chen J, Kelz MB, Zeng G, Steffen C, Shockett PE, Terwilliger G, Schatz DG, Nestler EJ. Inducible, reversible hair loss in transgenic mice. Transgenic Res 2002; 11:241-7. [PMID: 12113456 DOI: 10.1023/a:1015619604318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Telogen effluvium is a common type of hair loss. Although the morphological changes associated with telogen effluvium have been well characterized, the underlying molecular mechanisms remain unknown, and no animal models have been developed. We report here that inducible transgenic mice expressing high levels of the transcription factor, tTA (tetracycline transactivator), plus a reporter luciferase gene, show a reversible hair loss phenotype. Skin of these mice exhibits an increase in the number of hair follicles at the telogen phase, but a decreased number of follicles at the anagen phase. These changes resemble skin pathology seen in patients with telogen effluvium, which suggests that the inducible transgenic mice may be useful as a model for this disorder. Moreover, since overexpression of several other transgenes failed to cause skin pathology, the present findings also indicate types of molecular abnormalities that may cause reversible hair loss.
Collapse
Affiliation(s)
- Jingshan Chen
- Laboratory of Molecular Psychiatry, Yale University School of Medicine, Connecticut Mental Health Center, New Haven 06508, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang SQ, Tsiaras WG, Araki T, Wen G, Minichiello L, Klein R, Neel BG. Receptor-specific regulation of phosphatidylinositol 3'-kinase activation by the protein tyrosine phosphatase Shp2. Mol Cell Biol 2002; 22:4062-72. [PMID: 12024020 PMCID: PMC133866 DOI: 10.1128/mcb.22.12.4062-4072.2002] [Citation(s) in RCA: 212] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) play distinct roles in multiple biological systems. Many RTKs transmit similar signals, raising questions about how specificity is achieved. One potential mechanism for RTK specificity is control of the magnitude and kinetics of activation of downstream pathways. We have found that the protein tyrosine phosphatase Shp2 regulates the strength and duration of phosphatidylinositol 3'-kinase (PI3K) activation in the epidermal growth factor (EGF) receptor signaling pathway. Shp2 mutant fibroblasts exhibit increased association of the p85 subunit of PI3K with the scaffolding adapter Gab1 compared to that for wild-type (WT) fibroblasts or Shp2 mutant cells reconstituted with WT Shp2. Far-Western analysis suggests increased phosphorylation of p85 binding sites on Gab1. Gab1-associated PI3K activity is increased and PI3K-dependent downstream signals are enhanced in Shp2 mutant cells following EGF stimulation. Analogous results are obtained in fibroblasts inducibly expressing dominant-negative Shp2. Our results suggest that, in addition to its role as a positive component of the Ras-Erk pathway, Shp2 negatively regulates EGF-dependent PI3K activation by dephosphorylating Gab1 p85 binding sites, thereby terminating a previously proposed Gab1-PI3K positive feedback loop. Activation of PI3K-dependent pathways following stimulation by other growth factors is unaffected or decreased in Shp2 mutant cells. Thus, Shp2 regulates the kinetics and magnitude of RTK signaling in a receptor-specific manner.
Collapse
Affiliation(s)
- Si Qing Zhang
- Cancer Biology Program, Division of Hematology-Oncology, Department of Medicine, Beth Israel-Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|