1
|
Modder M, Coomans CP, Saaltink DJ, Tersteeg MMH, Hoogduin J, Scholten L, Pronk ACM, Lalai RA, Boelen A, Kalsbeek A, Rensen PCN, Vreugdenhil E, Kooijman S. Doublecortin-like knockdown in mice attenuates obesity by stimulating energy expenditure in adipose tissue. Sci Rep 2024; 14:19517. [PMID: 39174821 PMCID: PMC11341836 DOI: 10.1038/s41598-024-70639-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024] Open
Abstract
Crosstalk between peripheral metabolic organs and the central nervous system is essential for body weight control. At the base of the hypothalamus, β-tanycytes surround the portal capillaries and function as gatekeepers to facilitate transfer of substances from the circulation into the cerebrospinal fluid and vice versa. Here, we investigated the role of the neuroplasticity gene doublecortin-like (DCL), highly expressed by β-tanycytes, in body weight control and whole-body energy metabolism. We demonstrated that DCL-knockdown through a doxycycline-inducible shRNA expression system prevents body weight gain by reducing adiposity in mice. DCL-knockdown slightly increased whole-body energy expenditure possibly as a result of elevated circulating thyroid hormones. In white adipose tissue (WAT) triglyceride uptake was increased while the average adipocyte cell size was reduced. At histological level we observed clear signs of browning, and thus increased thermogenesis in WAT. We found no indications for stimulated thermogenesis in brown adipose tissue (BAT). Altogether, we demonstrate an important, though subtle, role of tanycytic DCL in body weight control through regulation of energy expenditure, and specifically WAT browning. Elucidating mechanisms underlying the role of DCL in regulating brain-peripheral crosstalk further might identify new treatment targets for obesity.
Collapse
Affiliation(s)
- Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Claudia P Coomans
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Dirk-Jan Saaltink
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Mayke M H Tersteeg
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Janna Hoogduin
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Leonie Scholten
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Endocrine Laboratory, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Erno Vreugdenhil
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
- Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands.
| |
Collapse
|
2
|
Molenaars M, Mir H, Alvarez SW, Arivazhagan L, Rosselot C, Zhan D, Park CY, Garcia-Ocana A, Schmidt AM, Possemato R. Acute inhibition of iron-sulfur cluster biosynthesis disrupts metabolic flexibility in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608291. [PMID: 39229169 PMCID: PMC11370322 DOI: 10.1101/2024.08.19.608291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Iron-sulfur clusters (ISCs) are cell-essential cofactors present in ∼60 proteins including subunits of OXPHOS complexes I-III, DNA polymerases, and iron-sensing proteins. Dysfunctions in ISC biosynthesis are associated with anemias, neurodegenerative disorders, and metabolic diseases. To assess consequences of acute ISC inhibition in a whole body setting, we developed a mouse model in which key ISC biosynthetic enzyme NFS1 can be acutely and reversibly suppressed. Contrary to in vitro ISC inhibition and pharmacological OXPHOS suppression, global NFS1 inhibition rapidly enhances lipid utilization and decreases adiposity without affecting caloric intake and physical activity. ISC proteins decrease, including key proteins involved in OXPHOS (SDHB), lipoic acid synthesis (LIAS), and insulin mRNA processing (CDKAL1), causing acute metabolic inflexibility. Age-related metabolic changes decelerate loss of adiposity substantially prolonged survival of mice with NFS1 inhibition. Thus, the observation that ISC metabolism impacts organismal fuel choice will aid in understanding the mechanisms underlying ISC diseases with increased risk for diabetes. Graphical abstract Highlights Acute ISC inhibition leads to rapid loss of adiposity in miceMulti-metabolic pathway disruption upon ISC deficiency blocks energy storageNfs1 inhibition induces glucose dyshomeostasis due to ISC deficiency in β-cellsEnergy distress caused by inhibition of ISC synthesis is attenuated in aged mice.
Collapse
|
3
|
Hong W, Gong P, Pan X, Liu Y, Qi G, Qi C, Qin S. Krüppel-like factor 7 deficiency disrupts corpus callosum development and neuronal migration in the developing mouse cerebral cortex. Brain Pathol 2023; 33:e13186. [PMID: 37401095 PMCID: PMC10467035 DOI: 10.1111/bpa.13186] [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: 10/07/2022] [Accepted: 06/16/2023] [Indexed: 07/05/2023] Open
Abstract
Krüppel-like Factor 7 (KLF7) is a zinc finger transcription factor that has a critical role in cellular differentiation, tumorigenesis, and regeneration. Mutations in Klf7 are associated with autism spectrum disorder, which is characterized by neurodevelopmental delay and intellectual disability. Here we show that KLF7 regulates neurogenesis and neuronal migration during mouse cortical development. Conditional depletion of KLF7 in neural progenitor cells resulted in agenesis of the corpus callosum, defects in neurogenesis, and impaired neuronal migration in the neocortex. Transcriptomic profiling analysis indicated that KLF7 regulates a cohort of genes involved in neuronal differentiation and migration, including p21 and Rac3. These findings provide insights into our understanding of the potential mechanisms underlying neurological defects associated with Klf7 mutations.
Collapse
Affiliation(s)
- Wentong Hong
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Xinjie Pan
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Congcong Qi
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesFudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
4
|
Metz M, O'Hare J, Cheng B, Puchowicz M, Buettner C, Scherer T. Brain insulin signaling suppresses lipolysis in the absence of peripheral insulin receptors and requires the MAPK pathway. Mol Metab 2023; 73:101723. [PMID: 37100238 DOI: 10.1016/j.molmet.2023.101723] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 04/28/2023] Open
Abstract
OBJECTIVES Insulin's ability to counterbalance catecholamine-induced lipolysis defines insulin action in adipose tissue. Insulin suppresses lipolysis directly at the level of the adipocyte and indirectly through signaling in the brain. Here, we further characterized the role of brain insulin signaling in regulating lipolysis and defined the intracellular insulin signaling pathway required for brain insulin to suppress lipolysis. METHODS We used hyperinsulinemic clamp studies coupled with tracer dilution techniques to assess insulin's ability to suppress lipolysis in two different mouse models with inducible insulin receptor depletion in all tissues (IRΔWB) or restricted to peripheral tissues excluding the brain (IRΔPER). To identify the underlying signaling pathway required for brain insulin to inhibit lipolysis, we continuously infused insulin +/- a PI3K or MAPK inhibitor into the mediobasal hypothalamus of male Sprague Dawley rats and assessed lipolysis during clamps. RESULTS Genetic insulin receptor deletion induced marked hyperglycemia and insulin resistance in both IRΔPER and IRΔWB mice. However, the ability of insulin to suppress lipolysis was largely preserved in IRΔPER, but completely obliterated in IRΔWB mice indicating that insulin is still able to suppress lipolysis as long as brain insulin receptors are present. Blocking the MAPK, but not the PI3K pathway impaired the inhibition of lipolysis by brain insulin signaling. CONCLUSION Brain insulin is required for insulin to suppress adipose tissue lipolysis and depends on intact hypothalamic MAPK signaling.
Collapse
Affiliation(s)
- Matthäus Metz
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, 1090 Austria
| | - James O'Hare
- Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - Bob Cheng
- Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA
| | - Michelle Puchowicz
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, 44106 USA
| | - Christoph Buettner
- Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA; Department of Medicine, Rutgers University, New Brunswick, NJ, 08901 USA.
| | - Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, 1090 Austria; Department of Medicine, Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029 USA.
| |
Collapse
|
5
|
Coomans C, Saaltink DJ, Deboer T, Tersteeg M, Lanooij S, Schneider AF, Mulder A, van Minnen J, Jost C, Koster AJ, Vreugdenhil E. Doublecortin-like expressing astrocytes of the suprachiasmatic nucleus are implicated in the biosynthesis of vasopressin and influences circadian rhythms. Glia 2021; 69:2752-2766. [PMID: 34343377 PMCID: PMC9291169 DOI: 10.1002/glia.24069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/09/2022]
Abstract
We have recently identified a novel plasticity protein, doublecortin-like (DCL), that is specifically expressed in the shell of the mouse suprachiasmatic nucleus (SCN). DCL is implicated in neuroplastic events, such as neurogenesis, that require structural rearrangements of the microtubule cytoskeleton, enabling dynamic movements of cell bodies and dendrites. We have inspected DCL expression in the SCN by confocal microscopy and found that DCL is expressed in GABA transporter-3 (GAT3)-positive astrocytes that envelope arginine vasopressin (AVP)-expressing cells. To investigate the role of these DCL-positive astrocytes in circadian rhythmicity, we have used transgenic mice expressing doxycycline-induced short-hairpin (sh) RNA's targeting DCL mRNA (DCL knockdown mice). Compared with littermate wild type (WT) controls, DCL-knockdown mice exhibit significant shorter circadian rest-activity periods in constant darkness and adjusted significantly faster to a jet-lag protocol. As DCL-positive astrocytes are closely associated with AVP-positive cells, we analyzed AVP expression in DCL-knockdown mice and in their WT littermates by 3D reconstructions and transmission electron microscopy (TEM). We found significantly higher numbers of AVP-positive cells with increased volume and more intensity in DCL-knockdown mice. We found alterations in the numbers of dense core vesicle-containing neurons at ZT8 and ZT20 suggesting that the peak and trough of neuropeptide biosynthesis is dampened in DCL-knockdown mice compared to WT littermates. Together, our data suggest an important role for the astrocytic plasticity in the regulation of circadian rhythms and point to the existence of a specific DCL+ astrocyte-AVP+ neuronal network located in the dorsal SCN implicated in AVP biosynthesis.
Collapse
Affiliation(s)
- Claudia Coomans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk-Jan Saaltink
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom Deboer
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mayke Tersteeg
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne Lanooij
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne Fleur Schneider
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aat Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan van Minnen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolina Jost
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abraham J Koster
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erno Vreugdenhil
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Doublecortin-Like Is Implicated in Adult Hippocampal Neurogenesis and in Motivational Aspects to Escape from an Aversive Environment in Male Mice. eNeuro 2020; 7:ENEURO.0324-19.2020. [PMID: 32994174 PMCID: PMC7568604 DOI: 10.1523/eneuro.0324-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/02/2022] Open
Abstract
Doublecortin (DCX)-like (DCL) is a microtubule (MT)-associated protein (MAP) that is highly homologous to DCX and is crucially involved in embryonic neurogenesis. Here, we have investigated the in vivo role of DCL in adult hippocampal neurogenesis by generating transgenic mice producing inducible shRNA molecules that specifically target DCL but no other splice variants produced by the DCLK gene. DCL knock-down (DCL-KD) resulted in a significant increase in the number of proliferating BrdU+ cells in the subgranular zone (SGZ) 1 d after BrdU administration. However, the number of surviving newborn adult NeuN+/BrdU+ neurons are significantly decreased when inspected four weeks after BrdU administration suggesting a blockade of neuronal differentiation after DCL-KD. In line with this, we observed an increase in the number of proliferating cells, but a significant decrease in postmitotic DCX+ cells that are characterized by long dendrites spanning all dentate gyrus layers. Behavioral analysis showed that DCL-KD strongly extended the escape latency of mice on the circular hole board (CHB) but did not affect other aspects of this behavioral task. Together, our results indicate a function for DCL in adult neurogenesis and in the motivation to escape from an aversive environment. In contrast to DCX, its pivotal role in the maturation of postmitotic neuronal progenitor cells (NPCs) marks DCL as a genuine adult neurogenesis indicator in the hippocampus.
Collapse
|
7
|
Manjunath H, Zhang H, Rehfeld F, Han J, Chang TC, Mendell JT. Suppression of Ribosomal Pausing by eIF5A Is Necessary to Maintain the Fidelity of Start Codon Selection. Cell Rep 2019; 29:3134-3146.e6. [PMID: 31801078 PMCID: PMC6917043 DOI: 10.1016/j.celrep.2019.10.129] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/19/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Sequences within 5' UTRs dictate the site and efficiency of translation initiation. In this study, an unbiased screen designed to interrogate the 5' UTR-mediated regulation of the growth-promoting gene MYC unexpectedly revealed the ribosomal pause relief factor eIF5A as a regulator of translation initiation codon selection. Depletion of eIF5A enhances upstream translation within 5' UTRs across yeast and human transcriptomes, including on the MYC transcript, where this results in increased production of an N-terminally extended protein. Furthermore, ribosome profiling experiments established that the function of eIF5A as a suppressor of ribosomal pausing at sites of suboptimal peptide bond formation is conserved in human cells. We present evidence that proximal ribosomal pausing on a transcript triggers enhanced use of upstream suboptimal or non-canonical initiation codons. Thus, we propose that eIF5A functions not only to maintain efficient translation elongation in eukaryotic cells but also to maintain the fidelity of translation initiation.
Collapse
Affiliation(s)
- Hema Manjunath
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - He Zhang
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8821, USA; Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390-8821, USA
| | - Frederick Rehfeld
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Jaeil Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Tsung-Cheng Chang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Joshua T Mendell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
8
|
Ren M, Miller PC, Schlame M, Phoon CKL. A critical appraisal of the tafazzin knockdown mouse model of Barth syndrome: what have we learned about pathogenesis and potential treatments? Am J Physiol Heart Circ Physiol 2019; 317:H1183-H1193. [PMID: 31603701 DOI: 10.1152/ajpheart.00504.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pediatric heart failure remains poorly understood, distinct in many aspects from adult heart failure. Limited data point to roles of altered mitochondrial functioning and, in particular, changes in mitochondrial lipids, especially cardiolipin. Barth syndrome is a mitochondrial disorder caused by tafazzin mutations that lead to abnormal cardiolipin profiles. Patients are afflicted by cardiomyopathy, skeletal myopathy, neutropenia, and growth delay. A mouse model of Barth syndrome was developed a decade ago, which relies on a doxycycline-inducible short hairpin RNA to knock down expression of tafazzin mRNA (TAZKD). Our objective was to review published data from the TAZKD mouse to determine its contributions to our pathogenetic understanding of, and potential treatment strategies for, Barth syndrome. In regard to the clinical syndrome, the reported physiological, biochemical, and ultrastructural abnormalities of the mouse model mirror those in Barth patients. Using this model, the peroxisome proliferator-activated receptor pan-agonist bezafibrate has been suggested as potential therapy because it ameliorated the cardiomyopathy in TAZKD mice, while increasing mitochondrial biogenesis. A clinical trial is now underway to test bezafibrate in Barth syndrome patients. Thus the TAZKD mouse model of Barth syndrome has led to important insights into disease pathogenesis and therapeutic targets, which can potentially translate to pediatric heart failure.
Collapse
Affiliation(s)
- Mindong Ren
- Department of Anesthesiology, New York University School of Medicine, New York, New York.,Department of Cell Biology, New York University School of Medicine, New York, New York
| | - Paighton C Miller
- Department of Pediatrics, Division of Pediatric Cardiology, New York University School of Medicine, New York, New York
| | - Michael Schlame
- Department of Anesthesiology, New York University School of Medicine, New York, New York.,Department of Cell Biology, New York University School of Medicine, New York, New York
| | - Colin K L Phoon
- Department of Pediatrics, Division of Pediatric Cardiology, New York University School of Medicine, New York, New York
| |
Collapse
|
9
|
Matthes S, Mosienko V, Popova E, Rivalan M, Bader M, Alenina N. Targeted Manipulation of Brain Serotonin: RNAi-Mediated Knockdown of Tryptophan Hydroxylase 2 in Rats. ACS Chem Neurosci 2019; 10:3207-3217. [PMID: 30977636 DOI: 10.1021/acschemneuro.8b00635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tryptophan hydroxylase (TPH) is the rate-limiting enzyme in the biosynthesis of the biogenic monoamine serotonin (5-hydroxytryptamine, 5-HT). Two existing TPH isoforms are responsible for the generation of two distinct serotonergic systems in vertebrates. TPH1, predominantly expressed in the gastrointestinal tract and pineal gland, mediates 5-HT biosynthesis in non-neuronal tissues, while TPH2, mainly found in the raphe nuclei of the brain stem, is accountable for the production of 5-HT in the brain. Neuronal 5-HT is a key regulator of mood and behavior and its deficiency has been implicated in a variety of neuropsychiatric disorders, e.g., depression and anxiety. To gain further insights into the complexity of central 5-HT modulations of physiological and pathophysiological processes, a new transgenic rat model, allowing an inducible gene knockdown of Tph2, was established based on doxycycline-inducible shRNA-expression. Biochemical phenotyping revealed a functional knockdown of Tph2 mRNA expression following oral doxycycline administration, with subsequent reductions in the corresponding levels of TPH2 enzyme expression and activity. Transgenic rats showed also significantly decreased tissue levels of 5-HT and its degradation product 5-Hydroxyindoleacetic acid (5-HIAA) in the raphe nuclei, hippocampus, hypothalamus, and cortex, while peripheral 5-HT concentrations in the blood remained unchanged. In summary, this novel transgenic rat model allows inducible manipulation of 5-HT biosynthesis specifically in the brain and may help to elucidate the role of 5-HT in the pathophysiology of affective disorders.
Collapse
Affiliation(s)
- Susann Matthes
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Valentina Mosienko
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- College of Medicine and Health, Institute of Biomedical and Clinical Sciences, University of Exeter, Hatherly Building, Prince of Wales Rd., EX4 4PS Exeter, United Kingdom
| | - Elena Popova
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| | - Marion Rivalan
- Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany
- Institute of Translational Biomedicine, St. Petersburg State University, Saint Petersburg 199034, Russia
| |
Collapse
|
10
|
Gurumurthy CB, Lloyd KCK. Generating mouse models for biomedical research: technological advances. Dis Model Mech 2019; 12:dmm029462. [PMID: 30626588 PMCID: PMC6361157 DOI: 10.1242/dmm.029462] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the past decade, new methods and procedures have been developed to generate genetically engineered mouse models of human disease. This At a Glance article highlights several recent technical advances in mouse genome manipulation that have transformed our ability to manipulate and study gene expression in the mouse. We discuss how conventional gene targeting by homologous recombination in embryonic stem cells has given way to more refined methods that enable allele-specific manipulation in zygotes. We also highlight advances in the use of programmable endonucleases that have greatly increased the feasibility and ease of editing the mouse genome. Together, these and other technologies provide researchers with the molecular tools to functionally annotate the mouse genome with greater fidelity and specificity, as well as to generate new mouse models using faster, simpler and less costly techniques.
Collapse
Affiliation(s)
- Channabasavaiah B Gurumurthy
- Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68106-5915, USA
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, University of Nebraska Medical Center, Omaha, NE 68106-5915, USA
| | - Kevin C Kent Lloyd
- Department of Surgery, School of Medicine, University of California, Davis, CA 95618, USA
- Mouse Biology Program, University of California, Davis, CA 95618, USA
| |
Collapse
|
11
|
Merentie M, Rissanen R, Lottonen-Raikaslehto L, Huusko J, Gurzeler E, Turunen MP, Holappa L, Mäkinen P, Ylä-Herttuala S. Doxycycline modulates VEGF-A expression: Failure of doxycycline-inducible lentivirus shRNA vector to knockdown VEGF-A expression in transgenic mice. PLoS One 2018; 13:e0190981. [PMID: 29351307 PMCID: PMC5774698 DOI: 10.1371/journal.pone.0190981] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/22/2017] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A) is the master regulator of angiogenesis, vascular permeability and growth. However, its role in mature blood vessels is still not well understood. To better understand the role of VEGF-A in the adult vasculature, we generated a VEGF-A knockdown mouse model carrying a doxycycline (dox)-regulatable short hairpin RNA (shRNA) transgene, which silences VEGF-A. The aim was to find the critical level of VEGF-A reduction for vascular well-being in vivo. In vitro, the dox-inducible lentiviral shRNA vector decreased VEGF-A expression efficiently and dose-dependently in mouse endothelial cells and cardiomyocytes. In the generated transgenic mice plasma VEGF-A levels decreased shortly after the dox treatment but returned back to normal after two weeks. VEGF-A expression decreased shortly after the dox treatment only in some tissues. Surprisingly, increasing the dox exposure time and dose led to elevated VEGF-A expression in some tissues of both wildtype and knockdown mice, suggesting that dox itself has an effect on VEGF-A expression. When the effect of dox on VEGF-A levels was further tested in naïve/non-transduced cells, the dox administration led to a decreased VEGF-A expression in endothelial cells but to an increased expression in cardiomyocytes. In conclusion, the VEGF-A knockdown was achieved in a dox-regulatable fashion with a VEGF-A shRNA vector in vitro, but not in the knockdown mouse model in vivo. Dox itself was found to regulate VEGF-A expression explaining the unexpected results in mice. The effect of dox on VEGF-A levels might at least partly explain its previously reported beneficial effects on myocardial and brain ischemia. Also, this effect on VEGF-A should be taken into account in all studies using dox-regulated vectors.
Collapse
Affiliation(s)
- Mari Merentie
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riina Rissanen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Line Lottonen-Raikaslehto
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jenni Huusko
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Erika Gurzeler
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko P. Turunen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lari Holappa
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
- Heart Center, Kuopio University Hospital, Kuopio, Finland
- * E-mail:
| |
Collapse
|
12
|
Greene C, Kealy J, Humphries MM, Gong Y, Hou J, Hudson N, Cassidy LM, Martiniano R, Shashi V, Hooper SR, Grant GA, Kenna PF, Norris K, Callaghan CK, Islam MDN, O’Mara SM, Najda Z, Campbell SG, Pachter JS, Thomas J, Williams NM, Humphries P, Murphy KC, Campbell M. Dose-dependent expression of claudin-5 is a modifying factor in schizophrenia. Mol Psychiatry 2018; 23:2156-2166. [PMID: 28993710 PMCID: PMC6298981 DOI: 10.1038/mp.2017.156] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/22/2017] [Accepted: 06/07/2017] [Indexed: 11/12/2022]
Abstract
Schizophrenia is a neurodevelopmental disorder that affects up to 1% of the general population. Various genes show associations with schizophrenia and a very weak nominal association with the tight junction protein, claudin-5, has previously been identified. Claudin-5 is expressed in endothelial cells forming part of the blood-brain barrier (BBB). Furthermore, schizophrenia occurs in 30% of individuals with 22q11 deletion syndrome (22q11DS), a population who are haploinsufficient for the claudin-5 gene. Here, we show that a variant in the claudin-5 gene is weakly associated with schizophrenia in 22q11DS, leading to 75% less claudin-5 being expressed in endothelial cells. We also show that targeted adeno-associated virus-mediated suppression of claudin-5 in the mouse brain results in localized BBB disruption and behavioural changes. Using an inducible 'knockdown' mouse model, we further link claudin-5 suppression with psychosis through a distinct behavioural phenotype showing impairments in learning and memory, anxiety-like behaviour and sensorimotor gating. In addition, these animals develop seizures and die after 3-4 weeks of claudin-5 suppression, reinforcing the crucial role of claudin-5 in normal neurological function. Finally, we show that anti-psychotic medications dose-dependently increase claudin-5 expression in vitro and in vivo while aberrant, discontinuous expression of claudin-5 in the brains of schizophrenic patients post mortem was observed compared to age-matched controls. Together, these data suggest that BBB disruption may be a modifying factor in the development of schizophrenia and that drugs directly targeting the BBB may offer new therapeutic opportunities for treating this disorder.
Collapse
Affiliation(s)
- C Greene
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - J Kealy
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - M M Humphries
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - Y Gong
- 0000 0001 2355 7002grid.4367.6Division of Renal Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO USA
| | - J Hou
- 0000 0001 2355 7002grid.4367.6Division of Renal Diseases, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO USA
| | - N Hudson
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - L M Cassidy
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - R Martiniano
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - V Shashi
- 0000000100241216grid.189509.cDepartment of Pediatrics, Duke University Medical Center, Durham, NC USA
| | - S R Hooper
- 0000000122483208grid.10698.36Department of Allied Health Sciences, University of North Carolina School of Medicine, Chapel Hill, NC USA
| | - G A Grant
- 0000000419368956grid.168010.eDepartment of Neurosurgery, Stanford University School of Medicine, Stanford, CA USA
| | - P F Kenna
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - K Norris
- 0000 0001 0303 540Xgrid.5884.1Biosciences Department, Faculty of Health and Wellbeing, Biosciences and Chemistry, Sheffield Hallam University, Sheffield, UK
| | - C K Callaghan
- 0000 0004 1936 9705grid.8217.cTrinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland ,0000 0004 1936 9705grid.8217.cSchool of Psychology, Trinity College Dublin, Dublin, Ireland
| | - M dN Islam
- 0000 0004 1936 9705grid.8217.cTrinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland ,0000 0004 1936 9705grid.8217.cSchool of Psychology, Trinity College Dublin, Dublin, Ireland
| | - S M O’Mara
- 0000 0004 1936 9705grid.8217.cTrinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland ,0000 0004 1936 9705grid.8217.cSchool of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Z Najda
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - S G Campbell
- 0000 0001 0303 540Xgrid.5884.1Biosciences Department, Faculty of Health and Wellbeing, Biosciences and Chemistry, Sheffield Hallam University, Sheffield, UK
| | - J S Pachter
- 0000000419370394grid.208078.5Department of Cell Biology, University of Connecticut Health Center, Farmington, CT USA
| | - J Thomas
- 0000 0001 0807 5670grid.5600.3Department of Psychological Medicine and Neurology, MRC Centre in Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - N M Williams
- 0000 0001 0807 5670grid.5600.3Department of Psychological Medicine and Neurology, MRC Centre in Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - P Humphries
- 0000 0004 1936 9705grid.8217.cDepartment of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland
| | - K C Murphy
- 0000 0004 0488 7120grid.4912.eDepartment of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M Campbell
- Department of Genetics, Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
13
|
Lee KH, Oghamian S, Park JA, Kang L, Laird PW. The REMOTE-control system: a system for reversible and tunable control of endogenous gene expression in mice. Nucleic Acids Res 2017; 45:12256-12269. [PMID: 28981717 PMCID: PMC5716148 DOI: 10.1093/nar/gkx829] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/07/2017] [Indexed: 12/30/2022] Open
Abstract
We report here a robust, tunable, and reversible transcription control system for endogenous genes. The REMOTE-control system (Reversible Manipulation of Transcription at Endogenous loci) employs enhanced lac repression and tet activation systems. With this approach, we show in mouse embryonic stem cells that endogenous Dnmt1 gene transcription could be up- or downregulated in a tunable, inducible, and reversible manner across nearly two orders of magnitude. Transcriptional repression of Dnmt1 by REMOTE-control was potent enough to cause embryonic lethality in mice, reminiscent of a genetic knockout of Dnmt1 and could substantially suppress intestinal polyp formation when applied to an ApcMin model. Binding by the enhanced lac repressor was sufficiently tight to allow strong attenuation of transcriptional elongation, even at operators located many kilobases downstream of the transcription start site and to produce invariably tight repression of all of the strong viral/mammalian promoters tested. Our approach of targeting tet transcriptional activators to the endogenous Dnmt1 promoter resulted in robust upregulation of this highly expressed housekeeping gene. Our system provides exquisite control of the level, timing, and cell-type specificity of endogenous gene expression, and the potency and versatility of the system will enable high resolution in vivo functional analyses.
Collapse
Affiliation(s)
- Kwang-Ho Lee
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | | - Jin-A Park
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Liang Kang
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
14
|
Chandran V, Gao K, Swarup V, Versano R, Dong H, Jordan MC, Geschwind DH. Inducible and reversible phenotypes in a novel mouse model of Friedreich's Ataxia. eLife 2017; 6:e30054. [PMID: 29257745 PMCID: PMC5736353 DOI: 10.7554/elife.30054] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022] Open
Abstract
Friedreich's ataxia (FRDA), the most common inherited ataxia, is caused by recessive mutations that reduce the levels of frataxin (FXN), a mitochondrial iron binding protein. We developed an inducible mouse model of Fxn deficiency that enabled us to control the onset and progression of disease phenotypes by the modulation of Fxn levels. Systemic knockdown of Fxn in adult mice led to multiple phenotypes paralleling those observed in human patients across multiple organ systems. By reversing knockdown after clinical features appear, we were able to determine to what extent observed phenotypes represent reversible cellular dysfunction. Remarkably, upon restoration of near wild-type FXN levels, we observed significant recovery of function, associated pathology and transcriptomic dysregulation even after substantial motor dysfunction and pathology were observed. This model will be of broad utility in therapeutic development and in refining our understanding of the relative contribution of reversible cellular dysfunction at different stages in disease.
Collapse
Affiliation(s)
- Vijayendran Chandran
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Kun Gao
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Vivek Swarup
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Revital Versano
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Hongmei Dong
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Maria C Jordan
- Department of Physiology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
15
|
Huang PY, Kandyba E, Jabouille A, Sjolund J, Kumar A, Halliwill K, McCreery M, DelRosario R, Kang HC, Wong CE, Seibler J, Beuger V, Pellegrino M, Sciambi A, Eastburn DJ, Balmain A. Lgr6 is a stem cell marker in mouse skin squamous cell carcinoma. Nat Genet 2017; 49:1624-1632. [PMID: 28945253 PMCID: PMC5662105 DOI: 10.1038/ng.3957] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/25/2017] [Indexed: 12/11/2022]
Abstract
The G-protein-coupled receptors Lgr4/5/6 are Wnt signalling mediators, but their functions in squamous carcinomas (SCCs) are unclear. Using lineage tracing in Lgr5-EGFP-CreERT2- and Lgr6-EGFP-CreERT2- Rosa26/Tomato reporter mice, we demonstrate that Lgr6, but not Lgr5, acts as an epithelial stem cell marker in vivo in SCCs. We identify, by single molecule in situ hybridisation and cell sorting, rare Lgr6-positive cells in immortalised keratinocytes, and show that their frequency increases in advanced SCCs. Lgr6 expression is enriched in cells with stem cell characteristics, and Lgr6 downregulation in vivo causes increased epidermal proliferation, with expanded lineage tracing from Lgr6+ epidermal stem cells. Surprisingly, Lgr6 germline knockout mice are predisposed to SCC development, by a mechanism that includes compensatory upregulation of Lgr5. These data provide a model for human patients with germline loss of function mutations in WNT pathway genes RSPO1 or LGR4, who show increased susceptibility to squamous tumour development.
Collapse
Affiliation(s)
- Phillips Y Huang
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA.,Genome Institute of Singapore, Singapore
| | - Eve Kandyba
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA
| | - Arnaud Jabouille
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA
| | - Jonas Sjolund
- Division of Translational Cancer Research, University of Lund, Lund, Sweden
| | - Atul Kumar
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA
| | - Kyle Halliwill
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA
| | - Melissa McCreery
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA
| | - Reyno DelRosario
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA
| | | | | | | | | | | | - Adam Sciambi
- Mission Bio, Inc., San Francisco, California, USA
| | | | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, California, USA.,Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
16
|
Zaccagnini G, Maimone B, Fuschi P, Maselli D, Spinetti G, Gaetano C, Martelli F. Overexpression of miR-210 and its significance in ischemic tissue damage. Sci Rep 2017; 7:9563. [PMID: 28842599 PMCID: PMC5573334 DOI: 10.1038/s41598-017-09763-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023] Open
Abstract
Hypoxia-induced miR-210 displays a pro-survival, cytoprotective and pro-angiogenic role in several in vitro systems. In vivo, we previously found that miR-210 inhibition increases ischemic damage. Here we describe the generation of a versatile transgenic mouse model allowing the evaluation of miR-210 therapeutic potential in ischemic cardiovascular diseases. We generated a Tet-On miR-210 transgenic mouse strain (TG-210) by targeted transgenesis in the ROSA26 locus. To functionally validate miR-210 transgenic mice, hindlimb ischemia was induced by femoral artery dissection. Blood perfusion was evaluated by power Doppler while tissue damage and inflammation were assessed by histological evaluation. We found that miR-210 levels were rapidly increased in TG-210 mice upon doxycycline administration. miR-210 overexpression was maintained over time and remained within physiological levels in multiple tissues. When hindlimb ischemia was induced, miR-210 overexpression protected from both muscular and vascular ischemic damage, decreased inflammatory cells density and allowed to maintain a better calf perfusion. In conclusion, we generated and functionally validated a miR-210 transgenic mouse model. Albeit validated in the context of a specific cardiovascular ischemic disease, miR-210 transgenic mice may also represent a useful model to assess the function of miR-210 in other physio-pathological conditions.
Collapse
Affiliation(s)
- G Zaccagnini
- Laboratory of Molecular Cardiology, Policlinico San Donato-IRCCS, 20097 San Donato Milanese, Milan, Italy
| | - B Maimone
- Laboratory of Molecular Cardiology, Policlinico San Donato-IRCCS, 20097 San Donato Milanese, Milan, Italy
| | - P Fuschi
- Laboratory of Molecular Cardiology, Policlinico San Donato-IRCCS, 20097 San Donato Milanese, Milan, Italy
| | - D Maselli
- Laboratory of Cardiovascular Research, MultiMedica-IRCCS, 20138, Milan, Italy
| | - G Spinetti
- Laboratory of Cardiovascular Research, MultiMedica-IRCCS, 20138, Milan, Italy
| | - C Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Internal Medicine Clinic III, Goethe University, Frankfurt am Main, Germany
| | - F Martelli
- Laboratory of Molecular Cardiology, Policlinico San Donato-IRCCS, 20097 San Donato Milanese, Milan, Italy.
| |
Collapse
|
17
|
Hastings NB, Wang X, Song L, Butts BD, Grotz D, Hargreaves R, Fred Hess J, Hong KLK, Huang CRR, Hyde L, Laverty M, Lee J, Levitan D, Lu SX, Maguire M, Mahadomrongkul V, McEachern EJ, Ouyang X, Rosahl TW, Selnick H, Stanton M, Terracina G, Vocadlo DJ, Wang G, Duffy JL, Parker EM, Zhang L. Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener 2017; 12:39. [PMID: 28521765 PMCID: PMC5437664 DOI: 10.1186/s13024-017-0181-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/09/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Hyperphosphorylation of microtubule-associated protein tau is a distinct feature of neurofibrillary tangles (NFTs) that are the hallmark of neurodegenerative tauopathies. O-GlcNAcylation is a lesser known post-translational modification of tau that involves the addition of N-acetylglucosamine onto serine and threonine residues. Inhibition of O-GlcNAcase (OGA), the enzyme responsible for the removal of O-GlcNAc modification, has been shown to reduce tau pathology in several transgenic models. Clarifying the underlying mechanism by which OGA inhibition leads to the reduction of pathological tau and identifying translatable measures to guide human dosing and efficacy determination would significantly facilitate the clinical development of OGA inhibitors for the treatment of tauopathies. METHODS Genetic and pharmacological approaches are used to evaluate the pharmacodynamic response of OGA inhibition. A panel of quantitative biochemical assays is established to assess the effect of OGA inhibition on pathological tau reduction. A "click" chemistry labeling method is developed for the detection of O-GlcNAcylated tau. RESULTS Substantial (>80%) OGA inhibition is required to observe a measurable increase in O-GlcNAcylated proteins in the brain. Sustained and substantial OGA inhibition via chronic treatment with Thiamet G leads to a significant reduction of aggregated tau and several phosphorylated tau species in the insoluble fraction of rTg4510 mouse brain and total tau in cerebrospinal fluid (CSF). O-GlcNAcylated tau is elevated by Thiamet G treatment and is found primarily in the soluble 55 kD tau species, but not in the insoluble 64 kD tau species thought as the pathological entity. CONCLUSION The present study demonstrates that chronic inhibition of OGA reduces pathological tau in the brain and total tau in the CSF of rTg4510 mice, most likely by directly increasing O-GlcNAcylation of tau and thereby maintaining tau in the soluble, non-toxic form by reducing tau aggregation and the accompanying panoply of deleterious post-translational modifications. These results clarify some conflicting observations regarding the effects and mechanism of OGA inhibition on tau pathology, provide pharmacodynamic tools to guide human dosing and identify CSF total tau as a potential translational biomarker. Therefore, this study provides additional support to develop OGA inhibitors as a treatment for Alzheimer's disease and other neurodegenerative tauopathies.
Collapse
Affiliation(s)
| | - Xiaohai Wang
- Department of In Vivo Pharmacology, West Point, PA USA
| | - Lixin Song
- Department of Neuroscience, Kenilworth, NJ USA
| | | | - Diane Grotz
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Kenilworth, NJ USA
| | | | | | | | | | - Lynn Hyde
- Department of Neuroscience, Kenilworth, NJ USA
| | | | - Julie Lee
- Department of Neuroscience, Kenilworth, NJ USA
| | - Diane Levitan
- Department of Molecular Biomarkers, Kenilworth, NJ USA
| | | | | | | | | | | | | | - Harold Selnick
- Discovery Chemistry, Merck Research Laboratories, West Point, PA USA
| | | | | | | | | | - Joseph L. Duffy
- Discovery Chemistry Merck Research Laboratories, Kenilworth, NJ USA
| | | | - Lili Zhang
- Department of Neuroscience, Kenilworth, NJ USA
| |
Collapse
|
18
|
Murine model and mechanisms of treatment-induced painful diabetic neuropathy. Neuroscience 2017; 354:136-145. [PMID: 28476321 DOI: 10.1016/j.neuroscience.2017.04.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 11/21/2022]
Abstract
Diabetes mellitus represents a group of metabolic diseases that are characterized by hyperglycemia caused by either lack of insulin production or a reduced ability to respond to insulin. It is estimated that there were 347 million people worldwide who suffered from diabetes in 2008 and incidence is predicted to double by 2050. Neuropathy is the most common complication of long-term diabetes and approximately 30% of these subjects develop chronic neuropathic pain. A distinct acute, severe form of neuropathic pain, called insulin neuritis or treatment-induced painful neuropathy of diabetes (TIND), may also occur shortly after initiation of intensive glycemic control, with an incidence rate of up to 10.9%. The pathological mechanisms leading to TIND, which is mostly unresponsive to analgesics, are not yet understood, impeding the development of therapies. Studies to date have been clinical and with limited cohorts of patients. In the current study, we developed chronic and acute insulin-induced neuropathic pain in mice with type 2 insulin-resistant diabetes. Furthermore, we determined that insulin-induced acute allodynia is independent of glycemia levels, can also be induced with Insulin-like Growth Factor 1 (IGF1) and be prevented by inhibition of AKT, providing evidence of an insulin/IGF1 signaling pathway-based mechanism for TIND. This mouse model is useful for the elucidation of mechanisms contributing to TIND and for the testing of new therapeutic approaches to treat TIND.
Collapse
|
19
|
Huang Y, Powers C, Moore V, Schafer C, Ren M, Phoon CKL, James JF, Glukhov AV, Javadov S, Vaz FM, Jefferies JL, Strauss AW, Khuchua Z. The PPAR pan-agonist bezafibrate ameliorates cardiomyopathy in a mouse model of Barth syndrome. Orphanet J Rare Dis 2017; 12:49. [PMID: 28279226 PMCID: PMC5345250 DOI: 10.1186/s13023-017-0605-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/27/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The PGC-1α/PPAR axis has been proposed as a potential therapeutic target for several metabolic disorders. The aim was to evaluate the efficacy of the pan-PPAR agonist, bezafibrate, in tafazzin knockdown mice (TazKD), a mouse model of Barth syndrome that exhibits age-dependent dilated cardiomyopathy with left ventricular (LV) dysfunction. RESULTS The effect of bezafibrate on cardiac function was evaluated by echocardiography in TazKD mice with or without beta-adrenergic stress. Adrenergic stress by chronic isoproterenol infusion exacerbates the cardiac phenotype in TazKD mice, significantly depressing LV systolic function by 4.5 months of age. Bezafibrate intake over 2 months substantially ameliorates the development of LV systolic dysfunction in isoproterenol-stressed TazKD mice. Without beta-adrenergic stress, TazKD mice develop dilated cardiomyopathy by 7 months of age. Prolonged treatment with suprapharmacological dose of bezafibrate (0.5% in rodent diet) over a 4-month period effectively prevented LV dilation in mice isoproterenol treatment. Bezafibrate increased mitochondrial biogenesis, however also promoted oxidative stress in cardiomyocytes. Surprisingly, improvement of systolic function in bezafibrate-treated mice was accompanied with simultaneous reduction of cardiolipin content and increase of monolysocardiolipin levels in cardiac muscle. CONCLUSIONS Thus, we demonstrate that bezafibrate has a potent therapeutic effect on preventing cardiac dysfunction in a mouse model of Barth syndrome with obvious implications for treating the human disease. Additional studies are needed to assess the potential benefits of PPAR agonists in humans with Barth syndrome.
Collapse
Affiliation(s)
- Yan Huang
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Corey Powers
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Victoria Moore
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Caitlin Schafer
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Mindong Ren
- Departments of Anesthesiology and Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Colin K L Phoon
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
| | - Jeanne F James
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Alexander V Glukhov
- Department of Biochemistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Frédéric M Vaz
- Academic Medical Center, Department of Clinical Chemistry and Pediatrics, Laboratory of Genetic Metabolic Disease (F0-224), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - John L Jefferies
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Arnold W Strauss
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA
| | - Zaza Khuchua
- The Heart Institute, Department of Pediatrics, the University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229-7020, USA.
| |
Collapse
|
20
|
Lu S, Li J, Lui KO. Individual Variation in Conditional β Cell Ablation Mice Contributes Significant Biases in Evaluating β Cell Functional Recovery. Front Endocrinol (Lausanne) 2017; 8:242. [PMID: 28959236 PMCID: PMC5604075 DOI: 10.3389/fendo.2017.00242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/31/2017] [Indexed: 11/20/2022] Open
Abstract
Despite the βDTA (Ins2-rtTA; Tet-DTA) mice have been developed as a valuable tool to study β cell regeneration, their individual variation in therapeutic efficacy has not been characterized. Here, we demonstrated that the βDTA mice exhibited significant variations in both spontaneous and acquired β cell regeneration. We found that doxycycline (DOX)-induced β cell death was sufficient to cause polydipsia, translating even subtle difference in drinking habit into large variations in actual DOX intake among individuals within the same group. Accumulating evidence shows that transient expression of VEGFA enhances β cell functional recovery after injury. Therefore, we utilized the chemically modified mRNA (modRNA) technology to enable transient yet efficient VEGFA expression in the pancreas after DOX-induced β cell death. Surprisingly, under optimized DOX dose permissive of β cell regeneration, VEGFA modRNA only demonstrated marginal benefits on β cell functional recovery with large individual variations. We also revealed that the therapeutic efficacy of VEGFA modRNA on β cell regeneration was dependent on the degree of β cell loss induced by the accumulated DOX intake. Therefore, our results highlight a significant contribution of individual variation in the βDTA model and call for attention in evaluating potential efficacy of therapeutic agents in β cell regeneration studies.
Collapse
Affiliation(s)
- Song Lu
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Jiatao Li
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Kathy O. Lui
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
- *Correspondence: Kathy O. Lui,
| |
Collapse
|
21
|
Abstract
The generation of a new genetically modified mouse strain is a big hurdle to take for many researchers. It is often unclear which steps and decisions have to be made prior to obtaining the desired mouse model. This review aims to help researchers by providing a decision guide that answers the essential questions that need to be asked before generating the most suitable genetically modified mouse line in the most optimal timeframe. The review includes the latest technologies in both the stem cell culture and gene editing tools, particularly CRISPR/Cas9, and provides compatibility guidelines for selecting among the different types of genetic modifications that can be introduced in the mouse genome and the various routes for introducing these modifications into the mouse germline.
Collapse
Affiliation(s)
- Ivo J Huijbers
- Mouse Clinic for Cancer and Aging, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Lin J, Liu Y, Zhan Y, Zhuang C, Liu L, Fu X, Xu W, Li J, Chen M, Cai Z, Huang W. Synthetic Tet-inducible small hairpin RNAs targeting hTERT or Bcl-2 inhibit malignant phenotypes of bladder cancer T24 and 5637 cells. Tumour Biol 2015; 37:3115-21. [PMID: 26427661 DOI: 10.1007/s13277-015-4122-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/20/2015] [Indexed: 02/05/2023] Open
Abstract
Small hairpin RNA (shRNA) can inhibit the malignant phenotypes of tumor cell through ribonucleic acid interference (RNAi). However, it is hardly to be regulated and it may induce few phenotypic changes. Here, we build a type of tetracycline (Tet)-inducible vectors which can achieve regulatable expression of shRNA in a time-dependent manner by using synthetic biology approach. In order to prove the effectiveness of this device, we chose hTERT and Bcl-2 as target genes and test the utility of the device on 5637 and T24 cell lines. The experiments show that the Tet-inducible small hairpin RNA can effectively suppress their target genes and generate anti-cancer effects on both 5637 and T24 cell lines. The device we build not only can inhibit proliferation but also can induce apoptosis and suppress migration of the bladder cancer cell lines 5637 and T24. The Tet-inducible small hairpin RNAs may provide a novel strategy for the treatment of human bladder cancer in the future.
Collapse
Affiliation(s)
- Junhao Lin
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
- Shantou University Medical College, Shantou, 515041, China.
| | - Yuchen Liu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yonghao Zhan
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- Shantou University Medical College, Shantou, 515041, China
| | - Chengle Zhuang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- Shantou University Medical College, Shantou, 515041, China
| | - Li Liu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- Shantou University Medical College, Shantou, 515041, China
| | - Xing Fu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Wen Xu
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jianfa Li
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
- Shantou University Medical College, Shantou, 515041, China
| | - Mingwei Chen
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Zhiming Cai
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
- Shantou University Medical College, Shantou, 515041, China.
| | - Weiren Huang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
- Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
23
|
Carty N, Berson N, Tillack K, Thiede C, Scholz D, Kottig K, Sedaghat Y, Gabrysiak C, Yohrling G, von der Kammer H, Ebneth A, Mack V, Munoz-Sanjuan I, Kwak S. Characterization of HTT inclusion size, location, and timing in the zQ175 mouse model of Huntington's disease: an in vivo high-content imaging study. PLoS One 2015; 10:e0123527. [PMID: 25859666 PMCID: PMC4393127 DOI: 10.1371/journal.pone.0123527] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/18/2015] [Indexed: 11/18/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Major pathological hallmarks of HD include inclusions of mutant huntingtin (mHTT) protein, loss of neurons predominantly in the caudate nucleus, and atrophy of multiple brain regions. However, the early sequence of histological events that manifest in region- and cell-specific manner has not been well characterized. Here we use a high-content histological approach to precisely monitor changes in HTT expression and characterize deposition dynamics of mHTT protein inclusion bodies in the recently characterized zQ175 knock-in mouse line. We carried out an automated multi-parameter quantitative analysis of individual cortical and striatal cells in tissue slices from mice aged 2-12 months and confirmed biochemical reports of an age-associated increase in mHTT inclusions in this model. We also found distinct regional and subregional dynamics for inclusion number, size and distribution with subcellular resolution. We used viral-mediated suppression of total HTT in the striatum of zQ175 mice as an example of a therapeutically-relevant but heterogeneously transducing strategy to demonstrate successful application of this platform to quantitatively assess target engagement and outcome on a cellular basis.
Collapse
Affiliation(s)
| | | | | | | | - Diana Scholz
- Evotec AG, Manfred Eigen Campus, Hamburg, Germany
| | | | | | | | - George Yohrling
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States of America
| | | | | | - Volker Mack
- Evotec AG, Manfred Eigen Campus, Hamburg, Germany
| | | | - Seung Kwak
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
24
|
Wu M, Dai G, Yao J, Hoyt S, Wang L, Mu J. Potentiation of insulin-mediated glucose lowering without elevated hypoglycemia risk by a small molecule insulin receptor modulator. PLoS One 2015; 10:e0122012. [PMID: 25799496 PMCID: PMC4370409 DOI: 10.1371/journal.pone.0122012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 02/10/2015] [Indexed: 12/30/2022] Open
Abstract
Insulin resistance is the key feature of type 2 diabetes and is manifested as attenuated insulin receptor (IR) signaling in response to same levels of insulin binding. Several small molecule IR activators have been identified and reported to exhibit insulin sensitization properties. One of these molecules, TLK19781 (Cmpd1), was investigated to examine its IR sensitizing action in vivo. Our data demonstrate that Cmpd1, at doses that produced minimal efficacy in the absence of insulin, potentiated insulin action during an OGTT in non-diabetic mice and enhanced insulin-mediated glucose lowering in diabetic mice. Interestingly, different from insulin alone, Cmpd1 combined with insulin showed enhanced efficacy and duration of action without increased hypoglycemia. To explore the mechanism underlying the apparent glucose dependent efficacy, tissue insulin signaling was compared in healthy and diabetic mice. Cmpd1 enhanced insulin’s effects on IR phosphorylation in both healthy and diabetic mice. In contrast, the compound potentiated insulin’s effects on Akt phosphorylation in diabetic but not in non-diabetic mice. These differential effects on signaling corresponding to glucose levels could be part of the mechanism for reduced hypoglycemia risk. The in vivo efficacy of Cmpd1 is specific and dependent on IR expression. Results from these studies support the idea of targeting IR for insulin sensitization, which carries low hypoglycemia risk by standalone treatment and could improve the effectiveness of insulin therapies.
Collapse
Affiliation(s)
- Margaret Wu
- Early Development and Discovery Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, NJ 08889, United States of America
| | - Ge Dai
- Early Development and Discovery Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, NJ 08889, United States of America
| | - Jun Yao
- Early Development and Discovery Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, NJ 08889, United States of America
| | - Scott Hoyt
- Early Development and Discovery Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, NJ 08889, United States of America
| | - Liangsu Wang
- Early Development and Discovery Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, NJ 08889, United States of America
| | - James Mu
- Early Development and Discovery Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, NJ 08889, United States of America
- * E-mail:
| |
Collapse
|
25
|
Abstract
RNA interference (RNAi) was discovered as a cellular defense mechanism more than decade ago. It has been exploited as a powerful tool for genetic manipulation. Characterized with specifically silencing target gene expression, it has great potential application for disease treatment. Currently, there are human clinical trials in progress or planned. Despite the excitement regarding this prominent technology, there are many obstacles and concerns that prevent RNAi from being widely used in the therapeutic field. Among them, the non-spatial and non-temporal control is the most difficult challenge, as well as off-target effects and triggering type I immune responses. Inducible RNAi technology can effectively regulate target genes by inducer-mediated small hairpin RNA expression. Combination with inducible regulation systems this makes RNAi technology more sophisticated and may provide a wider application field. This review discusses approaches of inducible RNAi systems, the potential problem areas and solutions and their therapeutic applications. Given the limitations discussed herein being resolved, we believe that inducible RNAi will be a major therapeutic modality within the next several years.
Collapse
Affiliation(s)
- Yi Liao
- a Key Laboratory of Biorheological Science and Technology , Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| | - Liling Tang
- a Key Laboratory of Biorheological Science and Technology , Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| |
Collapse
|
26
|
Lee H. Genetically engineered mouse models for drug development and preclinical trials. Biomol Ther (Seoul) 2014; 22:267-74. [PMID: 25143803 PMCID: PMC4131519 DOI: 10.4062/biomolther.2014.074] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/21/2022] Open
Abstract
Drug development and preclinical trials are challenging processes and more than 80% to 90% of drug candidates fail to gain approval from the United States Food and Drug Administration. Predictive and efficient tools are required to discover high quality targets and increase the probability of success in the process of new drug development. One such solution to the challenges faced in the development of new drugs and combination therapies is the use of low-cost and experimentally manageable in vivo animal models. Since the 1980's, scientists have been able to genetically modify the mouse genome by removing or replacing a specific gene, which has improved the identification and validation of target genes of interest. Now genetically engineered mouse models (GEMMs) are widely used and have proved to be a powerful tool in drug discovery processes. This review particularly covers recent fascinating technologies for drug discovery and preclinical trials, targeted transgenesis and RNAi mouse, including application and combination of inducible system. Improvements in technologies and the development of new GEMMs are expected to guide future applications of these models to drug discovery and preclinical trials.
Collapse
Affiliation(s)
- Ho Lee
- Division of Convergence Technology, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 410-769, Republic of Korea
| |
Collapse
|
27
|
Chan AWS. Progress and prospects for genetic modification of nonhuman primate models in biomedical research. ILAR J 2014; 54:211-23. [PMID: 24174443 DOI: 10.1093/ilar/ilt035] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The growing interest of modeling human diseases using genetically modified (transgenic) nonhuman primates (NHPs) is a direct result of NHPs (rhesus macaque, etc.) close relation to humans. NHPs share similar developmental paths with humans in their anatomy, physiology, genetics, and neural functions; and in their cognition, emotion, and social behavior. The NHP model within biomedical research has played an important role in the development of vaccines, assisted reproductive technologies, and new therapies for many diseases. Biomedical research has not been the primary role of NHPs. They have mainly been used for safety evaluation and pharmacokinetics studies, rather than determining therapeutic efficacy. The development of the first transgenic rhesus macaque (2001) revolutionized the role of NHP models in biomedicine. Development of the transgenic NHP model of Huntington's disease (2008), with distinctive clinical features, further suggested the uniqueness of the model system; and the potential role of the NHP model for human genetic disorders. Modeling human genetic diseases using NHPs will continue to thrive because of the latest advances in molecular, genetic, and embryo technologies. NHPs rising role in biomedical research, specifically pre-clinical studies, is foreseeable. The path toward the development of transgenic NHPs and the prospect of transgenic NHPs in their new role in future biomedicine needs to be reviewed. This article will focus on the advancement of transgenic NHPs in the past decade, including transgenic technologies and disease modeling. It will outline new technologies that may have significant impact in future NHP modeling and will conclude with a discussion of the future prospects of the transgenic NHP model.
Collapse
|
28
|
Michalak EM, Nacerddine K, Pietersen A, Beuger V, Pawlitzky I, Cornelissen-Steijger P, Wientjens E, Tanger E, Seibler J, van Lohuizen M, Jonkers J. Polycomb group gene Ezh2 regulates mammary gland morphogenesis and maintains the luminal progenitor pool. Stem Cells 2014; 31:1910-20. [PMID: 23712803 DOI: 10.1002/stem.1437] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/08/2013] [Accepted: 04/29/2013] [Indexed: 01/19/2023]
Abstract
Specification of the cellular hierarchy in the mammary gland involves complex signaling that remains poorly defined. Polycomb group proteins are known to contribute to the maintenance of stem cell identity through epigenetic modifications, leading to stable alterations in gene expression. The polycomb protein family member EZH2 is known to be important for stem cell maintenance in multiple tissues, but its role in mammary gland development and differentiation remains unknown. Our analyses show that EZH2 is predominantly expressed in luminal cells of the mouse mammary epithelium. As mammary gland development occurs mostly after birth, the analysis of EZH2 gene function in postnatal development is precluded by embryonic lethality of conventional EZH2 knockout mice. To investigate the role of EZH2 in normal mammary gland epithelium, we have generated novel transgenic mice that express doxycycline-regulatable short hairpin (sh) RNAs directed against Ezh2. Knockdown of EZH2 results in delayed outgrowth of the mammary epithelium during puberty, due to impaired terminal end bud formation and ductal elongation. Furthermore, our results demonstrate that EZH2 is required to maintain the luminal cell pool and may limit differentiation of luminal progenitors into CD61(+) differentiated luminal cells, suggesting a role for EZH2 in mammary luminal cell fate determination. Consistent with this, EZH2 knockdown reduced lobuloalveolar expansion during pregnancy, suggesting EZH2 is required for the differentiation of luminal progenitors to alveolar cells.
Collapse
Affiliation(s)
- Ewa Malgorzata Michalak
- Division of Molecular Pathology and Cancer Genomics Centre, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hematopoietic overexpression of FOG1 does not affect B-cells but reduces the number of circulating eosinophils. PLoS One 2014; 9:e92836. [PMID: 24747299 PMCID: PMC3991581 DOI: 10.1371/journal.pone.0092836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 02/26/2014] [Indexed: 12/31/2022] Open
Abstract
We have identified expression of the gene encoding the transcriptional coactivator FOG-1 (Friend of GATA-1; Zfpm1, Zinc finger protein multitype 1) in B lymphocytes. We found that FOG-1 expression is directly or indirectly dependent on the B cell-specific coactivator OBF-1 and that it is modulated during B cell development: expression is observed in early but not in late stages of B cell development. To directly test in vivo the role of FOG-1 in B lymphocytes, we developed a novel embryonic stem cell recombination system. For this, we combined homologous recombination with the FLP recombinase activity to rapidly generate embryonic stem cell lines carrying a Cre-inducible transgene at the Rosa26 locus. Using this system, we successfully generated transgenic mice where FOG-1 is conditionally overexpressed in mature B-cells or in the entire hematopoietic system. While overexpression of FOG-1 in B cells did not significantly affect B cell development or function, we found that enforced expression of FOG-1 throughout all hematopoietic lineages led to a reduction in the number of circulating eosinophils, confirming and extending to mammals the known function of FOG-1 in this lineage.
Collapse
|
30
|
Huijbers IJ, Bin Ali R, Pritchard C, Cozijnsen M, Kwon MC, Proost N, Song JY, de Vries H, Badhai J, Sutherland K, Krimpenfort P, Michalak EM, Jonkers J, Berns A. Rapid target gene validation in complex cancer mouse models using re-derived embryonic stem cells. EMBO Mol Med 2014; 6:212-25. [PMID: 24401838 PMCID: PMC3927956 DOI: 10.1002/emmm.201303297] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Human cancers modeled in Genetically Engineered Mouse Models (GEMMs) can provide important mechanistic insights into the molecular basis of tumor development and enable testing of new intervention strategies. The inherent complexity of these models, with often multiple modified tumor suppressor genes and oncogenes, has hampered their use as preclinical models for validating cancer genes and drug targets. In our newly developed approach for the fast generation of tumor cohorts we have overcome this obstacle, as exemplified for three GEMMs; two lung cancer models and one mesothelioma model. Three elements are central for this system; (i) The efficient derivation of authentic Embryonic Stem Cells (ESCs) from established GEMMs, (ii) the routine introduction of transgenes of choice in these GEMM-ESCs by Flp recombinase-mediated integration and (iii) the direct use of the chimeric animals in tumor cohorts. By applying stringent quality controls, the GEMM-ESC approach proofs to be a reliable and effective method to speed up cancer gene assessment and target validation. As proof-of-principle, we demonstrate that MycL1 is a key driver gene in Small Cell Lung Cancer.
Collapse
Affiliation(s)
- Ivo J Huijbers
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee-Kubli CA, Mixcoatl-Zecuatl T, Jolivalt CG, Calcutt NA. Animal models of diabetes-induced neuropathic pain. Curr Top Behav Neurosci 2014; 20:147-70. [PMID: 24510303 DOI: 10.1007/7854_2014_280] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neuropathy will afflict over half of the approximately 350 million people worldwide who currently suffer from diabetes and around one-third of diabetic patients with neuropathy will suffer from painful symptoms that may be spontaneous or stimulus evoked. Diabetes can be induced in rats or mice by genetic, dietary, or chemical means, and there are a variety of well-characterized models of diabetic neuropathy that replicate either type 1 or type 2 diabetes. Diabetic rodents display aspects of sensorimotor dysfunction such as stimulus-evoked allodynia and hyperalgesia that are widely used to model painful neuropathy. This allows investigation of pathogenic mechanisms and development of potential therapeutic interventions that may alleviate established pain or prevent onset of pain.
Collapse
|
32
|
Gerrits H, Paradé MCBC, Koonen-Reemst AMCB, Bakker NEC, Timmer-Hellings L, Sollewijn Gelpke MD, Gossen JA. Reversible infertility in a liver receptor homologue-1 (LRH-1)-knockdown mouse model. Reprod Fertil Dev 2014; 26:293-306. [DOI: 10.1071/rd12131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 12/22/2012] [Indexed: 12/15/2022] Open
Abstract
Liver receptor homologue-1 (LRH-1) is an orphan nuclear receptor that has been implicated in steroid hormone biosynthesis and fertility. Herein we describe a transgenic inducible short hairpin (sh) RNA mouse model that was used to study the effect of transient LRH-1 knockdown in vivo. Induction of expression of the shRNA directed against LRH-1 for 2–6 weeks resulted in 80% knockdown of LRH-1 protein in the ovary and complete infertility. Gonadotropin hyperstimulation could not rescue the observed defects in ovulation and corpus luteum formation in LRH-1-knockdown mice. The infertility phenotype was fully reversible because LRH-1-knockdown females became pregnant and delivered normal size litters and healthy pups after cessation of LRH-1 shRNA expression. Timed ovarian microarray analysis showed that, in line with the observed decrease in plasma progesterone levels, key steroid biosynthesis genes, namely Star, Cyp11a1, Hsd3b and Scarb1, were downregulated in LRH-1-knockdown ovaries. In contrast with what has been described previously, no clear effect was observed on oestrogenic activity in LRH-1-knockdown mice. Only Sult1e1 and, surprisingly, Hsd17b7 expression was modulated with potentially opposite effects on oestradiol bioavailability. In conclusion, the fully reversible infertility phenotype of LRH-1-knockdown mice shows the feasibility of an LRH-1 antagonist as new contraceptive therapy with a mechanism of action that most prominently affects cholesterol availability and progesterone production.
Collapse
|
33
|
Livshits G, Lowe SW. Accelerating cancer modeling with RNAi and nongermline genetically engineered mouse models. Cold Spring Harb Protoc 2013; 2013:2013/11/pdb.top069856. [PMID: 24184755 DOI: 10.1101/pdb.top069856] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
For more than two decades, genetically engineered mouse models have been key to our mechanistic understanding of tumorigenesis and cancer progression. Recently, the massive quantity of data emerging from cancer genomics studies has demanded a corresponding increase in the efficiency and throughput of in vivo models for functional testing of putative cancer genes. Already a mainstay of cancer research, recent innovations in RNA interference (RNAi) technology have extended its utility for studying gene function and genetic interactions, enabling tissue-specific, inducible and reversible gene silencing in vivo. Concurrent advances in embryonic stem cell (ESC) culture and genome engineering have accelerated several steps of genetically engineered mouse model production and have facilitated the incorporation of RNAi technology into these models. Here, we review the current state of these technologies and examine how their integration has the potential to dramatically enhance the throughput and capabilities of animal models for cancer.
Collapse
Affiliation(s)
- Geulah Livshits
- Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | | |
Collapse
|
34
|
Zhao B, Yang C, Yang S, Gao Y, Wang J. Construction of conditional lentivirus-mediated shRNA vector targeting the human Mirk gene and identification of RNAi efficiency in rhabdomyosarcoma RD cells. Int J Oncol 2013; 43:1253-9. [PMID: 23913162 DOI: 10.3892/ijo.2013.2048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 07/23/2013] [Indexed: 11/05/2022] Open
Abstract
Rhabdomyosarcoma is the most common malignant soft tissue tumor in children. It has been demonstrated that Mirk as an activated protein kinase is overexpressed in rhabdomyosarcoma cells, which may be correlated with tumorigenesis. The aim of the present study was to explore the possibility of Mirk gene as a therapeutic target for the treatment of rhabdomyosarcoma, and the use of RNA interference in a temporally and spatially restricted manner to study the function of the target gene would be highly beneficial. To address this problem, a conditional lentivirus-mediated short hairpin RNA targeting human Mirk gene was constructed and employed to reduce endogenous Mirk expression in the rhabdomyosarcoma RD cell line in vitro. The expression of Mirk shRNA in RD cells transduced with this recombinant vector could be tracked with the expression of red fluorescent protein by the administration of doxycycline. A stable transgenic RD line was generated by transducing RD lines with the packaging viral particles. Quantitative PCR and western blot analysis indicated that the mRNA and protein levels of Mirk in the transgenic RD cells were significantly lower compared to those in the controls. In addition, the increasing apoptosis of RD cells induced by silencing of the Mirk gene was also observed. Overall, the results demonstrated that this recombinant vector-based RNAi expression system is an efficient approach to knockdown Mirk gene expression in the rhabdomyosarcoma RD cell line, which could, thereby, provide both a protocol to study the role of Mirk gene in tumor cells and a safer gene therapy in the clinic.
Collapse
Affiliation(s)
- Boming Zhao
- Department of Orthopaedic Surgery, The No. 1 People's Hospital of Jingzhou, Jingzhou, P.R. China
| | | | | | | | | |
Collapse
|
35
|
Efficient ROSA26-Based Conditional and/or Inducible Transgenesis Using RMCE-Compatible F1 Hybrid Mouse Embryonic Stem Cells. Stem Cell Rev Rep 2013; 9:774-85. [DOI: 10.1007/s12015-013-9458-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
36
|
van der Weyden L, Adams DJ. Cancer of mice and men: old twists and new tails. J Pathol 2013; 230:4-16. [PMID: 23436574 DOI: 10.1002/path.4184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 01/28/2013] [Accepted: 02/16/2013] [Indexed: 12/18/2022]
Abstract
In this review we set out to celebrate the contribution that mouse models of human cancer have made to our understanding of the fundamental mechanisms driving tumourigenesis. We take the opportunity to look forward to how the mouse will be used to model cancer and the tools and technologies that will be applied, and indulge in looking back at the key advances the mouse has made possible.
Collapse
|
37
|
Focal segmental glomerulosclerosis is induced by microRNA-193a and its downregulation of WT1. Nat Med 2013; 19:481-7. [PMID: 23502960 DOI: 10.1038/nm.3142] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 02/16/2013] [Indexed: 02/08/2023]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a frequent and severe glomerular disease characterized by destabilization of podocyte foot processes. We report that transgenic expression of the microRNA miR-193a in mice rapidly induces FSGS with extensive podocyte foot process effacement. Mechanistically, miR-193a inhibits the expression of the Wilms' tumor protein (WT1), a transcription factor and master regulator of podocyte differentiation and homeostasis. Decreased expression levels of WT1 lead to downregulation of its target genes PODXL (podocalyxin) and NPHS1 (nephrin), as well as several other genes crucial for the architecture of podocytes, initiating a catastrophic collapse of the entire podocyte-stabilizing system. We found upregulation of miR-193a in isolated glomeruli from individuals with FSGS compared to normal kidneys or individuals with other glomerular diseases. Thus, upregulation of miR-193a provides a new pathogenic mechanism for FSGS and is a potential therapeutic target.
Collapse
|
38
|
In vivo knockdown of TAK1 accelerates bone marrow proliferation/differentiation and induces systemic inflammation. PLoS One 2013; 8:e57348. [PMID: 23505428 PMCID: PMC3591390 DOI: 10.1371/journal.pone.0057348] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/21/2013] [Indexed: 11/29/2022] Open
Abstract
TAK1 (TGF-β Activated Kinase 1) is a MAPK kinase kinase, which activates the p38- and JNK-MAPK and NF-κB pathways downstream of receptors such as Toll-Like-, cytokine- and T-cell and B-cell receptors. Representing such an important node in the pro-inflammatory signal-transduction network, the function of TAK1 has been studied extensively. TAK1 knock-out mice are embryonic lethal, while conditional knock-out mice demonstrated either a pro- or anti-inflammatory function. To study the function of TAK1 protein in the adult immune system, we generated and characterized a transgenic mouse expressing TAK1 shRNA under the control of a doxycycline-inducible promoter. Following treatment of TAK-1 shRNA transgenic mice with doxycycline an effective knockdown of TAK1 protein levels was observed in lymphoid organs and cells in the peritoneal cavity (>50% down regulation). TAK1 knockdown resulted in significant changes in leukocyte populations in blood, bone marrow, spleen and peritoneal cavity. Upon TAK1 knockdown mice demonstrated splenomegaly, signs of systemic inflammation (increased levels of circulating cytokines and increase in cellularity of the B-cell areas and in germinal center development in the follicles) and degenerative changes in heart, kidneys and liver. Not surprisingly, TAK1-Tg mice treated with LPS or anti-CD3 antibodies showed enhanced cytokine/chemokine secretion. Finally, analysis of progenitor cells in the bone marrow upon doxycycline treatment showed increased proliferation and differentiation of myeloid progenitor cells. Given the similarity of the phenotype with TGF-β genetic models, our data suggest that in our model the function of TAK1 in TGF-β signal-transduction is overruling its function in pro-inflammatory signaling.
Collapse
|
39
|
Abstract
Gene silencing by RNA interference (RNAi) has become a standard method for the characterization of gene function in mammalian cells. Short hairpin (sh) RNAs expressed from stably integrated vectors mediate gene knockdown both in cultured cells and in mice, presenting a fast alternative to gene knockout approaches. We describe three strategies to control gene silencing in mice that can be applied to any transcript of interest. This shRNA based approach enables either i) constitutive body-wide knockdown, ii) cell type-specific knockdown controlled by Cre recombinase, or iii) inducible body-wide knockdown controlled by doxycycline. For reliable expression the shRNA vector of interest is inserted into a Rosa26 docking site of ES cells by a site-specific recombinase. These ES cells can then be used to generate shRNA transgenic mice. This technology enables the production of adult knockdown mice within 11 months for an expedite in vivo validation of drug targets.
Collapse
|
40
|
MitoNEET-driven alterations in adipocyte mitochondrial activity reveal a crucial adaptive process that preserves insulin sensitivity in obesity. Nat Med 2012; 18:1539-49. [PMID: 22961109 DOI: 10.1038/nm.2899] [Citation(s) in RCA: 377] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 07/10/2012] [Indexed: 12/11/2022]
Abstract
We examined mouse models with altered adipocyte expression of mitoNEET, a protein residing in the mitochondrial outer membrane, to probe its impact on mitochondrial function and subsequent cellular responses. We found that overexpression of mitoNEET enhances lipid uptake and storage, leading to an expansion of the mass of adipose tissue. Despite the resulting massive obesity, benign aspects of adipose tissue expansion prevail, and insulin sensitivity is preserved. Mechanistically, we also found that mitoNEET inhibits mitochondrial iron transport into the matrix and, because iron is a rate-limiting component for electron transport, lowers the rate of β-oxidation. This effect is associated with a lower mitochondrial membrane potential and lower levels of reactive oxygen species-induced damage, along with increased production of adiponectin. Conversely, a reduction in mitoNEET expression enhances mitochondrial respiratory capacity through enhanced iron content in the matrix, ultimately corresponding to less weight gain on a high-fat diet. However, this reduction in mitoNEET expression also causes heightened oxidative stress and glucose intolerance. Thus, manipulation of mitochondrial function by varying mitoNEET expression markedly affects the dynamics of cellular and whole-body lipid homeostasis.
Collapse
|
41
|
Abstract
RNA interference (RNAi) is a promising strategy to suppress the expression of disease-relevant genes and induce post-transcriptional gene silencing. Their simplicity and stability endow RNAi with great advantages in molecular medicine. Several RNAi-based drugs are in various stages of clinical investigation. This review summarizes the ongoing research endeavors on RNAi in molecular medicine, delivery systems for RNAi-based drugs, and a compendium of RNAi drugs in different stages of clinical development. Of special interest are RNAi-based drug target discovery and validation, delivery systems for RNAi-based drugs, such as nanoparticles, rabies virus protein-based vehicles, and bacteriophages for RNA packaging.
Collapse
Affiliation(s)
- Jing Chen
- Institute of Modern, Biopharmaceuticals, State Key, Laboratory Breeding Base of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | | |
Collapse
|
42
|
Sprüssel A, Schulte JH, Weber S, Necke M, Händschke K, Thor T, Pajtler KW, Schramm A, König K, Diehl L, Mestdagh P, Vandesompele J, Speleman F, Jastrow H, Heukamp LC, Schüle R, Dührsen U, Buettner R, Eggert A, Göthert JR. Lysine-specific demethylase 1 restricts hematopoietic progenitor proliferation and is essential for terminal differentiation. Leukemia 2012; 26:2039-51. [PMID: 22699452 DOI: 10.1038/leu.2012.157] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lysine (K)-specific demethylase 1A (LSD1/KDM1A) has been identified as a potential therapeutic target in solid cancers and more recently in acute myeloid leukemia. However, the potential side effects of a LSD1-inhibitory therapy remain elusive. Here, we show, with a newly established conditional in vivo knockdown model, that LSD1 represents a central regulator of hematopoietic stem and progenitor cells. LSD1 knockdown (LSD1-kd) expanded progenitor numbers by enhancing their proliferative behavior. LSD1-kd led to an extensive expansion of granulomonocytic, erythroid and megakaryocytic progenitors. In contrast, terminal granulopoiesis, erythropoiesis and platelet production were severely inhibited. The only exception was monopoiesis, which was promoted by LSD1 deficiency. Importantly, we showed that peripheral blood granulocytopenia, monocytosis, anemia and thrombocytopenia were reversible after LSD1-kd termination. Extramedullary splenic hematopoiesis contributed to the phenotypic reversion, and progenitor populations remained expanded. LSD1-kd was associated with the upregulation of key hematopoietic genes, including Gfi1b, Hoxa9 and Meis1, which are known regulators of the HSC/progenitor compartment. We also demonstrated that LSD1-kd abrogated Gfi1b-negative autoregulation by crossing LSD1-kd with Gfi1b:GFP mice. Taken together, our findings distinguish LSD1 as a critical regulator of hematopoiesis and point to severe, but reversible, side effects of a LSD1-targeted therapy.
Collapse
Affiliation(s)
- A Sprüssel
- Department of Pediatric Oncology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Analyses of the human genome have proven extremely successful in identifying changes that contribute to human disease. Genetically engineered mice provide a powerful tool to analyze these changes, although they are slow and costly and do not always recapitulate human biology. Recent advances in genomic technologies, rodent-modeling approaches, and the production of patient-derived reprogrammed cell lines now provide a plethora of complementary systems to study disease states and test new therapies. Continued evolution and integration of these model systems will be the key to realizing the benefits of the genomic revolution and refining our understanding and treatment of human diseases.
Collapse
|
44
|
Phoon CKL, Acehan D, Schlame M, Stokes DL, Edelman-Novemsky I, Yu D, Xu Y, Viswanathan N, Ren M. Tafazzin knockdown in mice leads to a developmental cardiomyopathy with early diastolic dysfunction preceding myocardial noncompaction. J Am Heart Assoc 2012; 1:jah313. [PMID: 23130124 PMCID: PMC3487377 DOI: 10.1161/jaha.111.000455] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 02/21/2012] [Indexed: 11/16/2022]
Abstract
BACKGROUND Barth syndrome is a rare, multisystem disorder caused by mutations in tafazzin that lead to cardiolipin deficiency and mitochondrial abnormalities. Patients most commonly develop an early-onset cardiomyopathy in infancy or fetal life. METHODS AND RESULTS Knockdown of tafazzin (TAZKD) in a mouse model was induced from the start of gestation via a doxycycline-inducible shRNA transgenic approach. All liveborn TAZKD mice died within the neonatal period, and in vivo echocardiography revealed prenatal loss of TAZKD embryos at E12.5-14.5. TAZKD E13.5 embryos and newborn mice demonstrated significant tafazzin knockdown, and mass spectrometry analysis of hearts revealed abnormal cardiolipin profiles typical of Barth syndrome. Electron microscopy of TAZKD hearts demonstrated ultrastructural abnormalities in mitochondria at both E13.5 and newborn stages. Newborn TAZKD mice exhibited a significant reduction in total mitochondrial area, smaller size of individual mitochondria, reduced cristae density, and disruption of the normal parallel orientation between mitochondria and sarcomeres. Echocardiography of E13.5 and newborn TAZKD mice showed good systolic function, but early diastolic dysfunction was evident from an abnormal flow pattern in the dorsal aorta. Strikingly, histology of E13.5 and newborn TAZKD hearts showed myocardial thinning, hypertrabeculation and noncompaction, and defective ventricular septation. Altered cellular proliferation occurring within a narrow developmental window accompanied the myocardial hypertrabeculation-noncompaction. CONCLUSIONS In this murine model, tafazzin deficiency leads to a unique developmental cardiomyopathy characterized by ventricular myocardial hypertrabeculation-noncompaction and early lethality. A central role of cardiolipin and mitochondrial functioning is strongly implicated in cardiomyocyte differentiation and myocardial patterning required for heart development. (J Am Heart Assoc. 2012;1:jah3-e000455 doi: 10.1161/JAHA.111.000455.).
Collapse
Affiliation(s)
- Colin K L Phoon
- Department of Pediatrics (Pediatric Cardiology), New York University School of Medicine, New York (C.K.L.P., N.V.)
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The discovery of microRNAs (miRNAs) almost two decades ago established a new paradigm of gene regulation. During the past ten years these tiny non-coding RNAs have been linked to virtually all known physiological and pathological processes, including cancer. In the same way as certain key protein-coding genes, miRNAs can be deregulated in cancer, in which they can function as a group to mark differentiation states or individually as bona fide oncogenes or tumour suppressors. Importantly, miRNA biology can be harnessed experimentally to investigate cancer phenotypes or used therapeutically as a target for drugs or as the drug itself.
Collapse
|
46
|
Abstract
The discovery of microRNAs (miRNAs) almost two decades ago established a new paradigm of gene regulation. During the past ten years these tiny non-coding RNAs have been linked to virtually all known physiological and pathological processes, including cancer. In the same way as certain key protein-coding genes, miRNAs can be deregulated in cancer, in which they can function as a group to mark differentiation states or individually as bona fide oncogenes or tumour suppressors. Importantly, miRNA biology can be harnessed experimentally to investigate cancer phenotypes or used therapeutically as a target for drugs or as the drug itself.
Collapse
Affiliation(s)
- Amaia Lujambio
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | | |
Collapse
|
47
|
Abstract
Drug resistance is one of the most pressing problems in treating cancer patients today. Local and regional disease can usually be adequately treated, but patients eventually die from distant metastases that have become resistant to all available chemotherapy. Although work on cultured tumor cell lines has yielded a lot of information on potential drug resistance mechanisms, it has proven difficult to translate these results to clinical drug resistance in patients. The controversy regarding the contribution of ABC transporters to drug resistance in patients is one example. The study of genetically engineered mouse models (GEMMs), which closely resemble cancer in human patients, can help to bridge this gap. In models for BRCA1- or BRCA2-associated breast cancer, we observed a substantial synergy between the defect in homology-directed DNA repair and sensitivity to DNA-targeting drugs. Nevertheless, tumors are not easily eradicated and eventually drug resistance develops. In this review we will discuss the use of the new generation mouse models to address major clinical problems, such as mechanisms of drug resistance, predicting chemotherapy response or characterizing the nature of residual tumor cells that escape eradication. Moreover, we will address the contribution of ABC transporters to drug resistance in our model.
Collapse
Affiliation(s)
- Sven Rottenberg
- Division of Molecular Biology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| | | |
Collapse
|
48
|
Gama Sosa MA, De Gasperi R, Elder GA. Modeling human neurodegenerative diseases in transgenic systems. Hum Genet 2011; 131:535-63. [PMID: 22167414 DOI: 10.1007/s00439-011-1119-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 11/23/2011] [Indexed: 02/07/2023]
Abstract
Transgenic systems are widely used to study the cellular and molecular basis of human neurodegenerative diseases. A wide variety of model organisms have been utilized, including bacteria (Escherichia coli), plants (Arabidopsis thaliana), nematodes (Caenorhabditis elegans), arthropods (Drosophila melanogaster), fish (zebrafish, Danio rerio), rodents (mouse, Mus musculus and rat, Rattus norvegicus) as well as non-human primates (rhesus monkey, Macaca mulatta). These transgenic systems have enormous value for understanding the pathophysiological basis of these disorders and have, in some cases, been instrumental in the development of therapeutic approaches to treat these conditions. In this review, we discuss the most commonly used model organisms and the methodologies available for the preparation of transgenic organisms. Moreover, we provide selected examples of the use of these technologies for the preparation of transgenic animal models of neurodegenerative diseases, including Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) and Parkinson's disease (PD) and discuss the application of these technologies to AD as an example of how transgenic modeling has affected the study of human neurodegenerative diseases.
Collapse
Affiliation(s)
- Miguel A Gama Sosa
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA.
| | | | | |
Collapse
|
49
|
Scherer T, Buettner C. Yin and Yang of hypothalamic insulin and leptin signaling in regulating white adipose tissue metabolism. Rev Endocr Metab Disord 2011; 12:235-43. [PMID: 21713385 PMCID: PMC3253350 DOI: 10.1007/s11154-011-9190-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fatty acids released from white adipose tissue (WAT) provide important energy substrates during fasting. However, uncontrolled fatty acid release from WAT during non-fasting states causes lipotoxicity and promotes inflammation and insulin resistance, which can lead to and worsen type 2 diabetes (DM2). WAT is also a source for insulin sensitizing fatty acids such as palmitoleate produced during de novo lipogenesis. Insulin and leptin are two major hormonal adiposity signals that control energy homeostasis through signaling in the central nervous system. Both hormones have been implicated to regulate both WAT lipolysis and de novo lipogenesis through the mediobasal hypothalamus (MBH) in an opposing fashion independent of their respective peripheral receptors. Here, we review the current literature on brain leptin and insulin action in regulating WAT metabolism and discuss potential mechanisms and neuro-anatomical substrates that could explain the opposing effects of central leptin and insulin. Finally, we discuss the role of impaired hypothalamic control of WAT metabolism in the pathogenesis of insulin resistance, metabolic inflexibility and type 2 diabetes.
Collapse
Affiliation(s)
- Thomas Scherer
- Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1055, New York, NY 10029-6574, USA
| | - Christoph Buettner
- Department of Medicine and Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1055, New York, NY 10029-6574, USA
| |
Collapse
|
50
|
Castro-Perez JM, Roddy TP, Shah V, Wang SP, Ouyang X, Ogawa A, McLaren DG, Tadin-Strapps M, Robinson MJ, Bartz SR, Ason B, Chen Y, Previs SF, Wong KK, Vreeken RJ, Johns DG, Hubbard BK, Hankemeier T, Mitnaul L. Attenuation of Slc27a5 gene expression followed by LC-MS measurement of bile acid reconjugation using metabolomics and a stable isotope tracer strategy. J Proteome Res 2011; 10:4683-91. [PMID: 21819150 DOI: 10.1021/pr200475g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The purpose of this study was to evaluate the use of high resolution LC-MS together with metabolomics and D(4)-cholic acid (D(4)-CA) as a metabolic tracer to measure the metabolism and reconjugation of bile acids (BAs) in vitro and in vivo. Metabolic tracers are very important because they allow for the direct detection (substrate-to-product) of small and significant biological perturbations that may not be apparent when monitoring "static" endogenous levels of particular metabolites. Slc27a5, also known as fatty acid transport protein 5 (FATP5), is the hepatic BA-CoA ligase involved in reconjugating BAs during enterohepatic BA recycling. Using Slc27a5-cKD mice, silencing of ∼90% gene expression was achieved followed by reduction in the reconjugation of D(4)-CA to D(4)-taurocholic acid (D(4)-TCA), as well as other conjugated BA metabolites in plasma (p = 0.0031). The method described allowed a rapid measure of many D(4) and endogenous BA. Analysis of bile resulted in the detection of 39 BA metabolites from a 13 min analytical run. Finally, the utilization of a novel high resolution mass spectrometry method in combination with metabolomics and a stable isotope metabolic tracer allowed for the detection of targeted and untargeted BAs following silencing of the Slc27a5 gene in primary hepatocytes and in mice.
Collapse
|