152 results on '"Saunders TL"'
Search Results
2. A Requirement for Argonaute 4 in Mammalian Antiviral Defense
- Author
-
Adiliaghdam, F, Basavappa, M, Saunders, TL, Harjanto, D, Prior, JT, Cronkite, DA, Papavasiliou, N, Jeffrey, KL, Adiliaghdam, F, Basavappa, M, Saunders, TL, Harjanto, D, Prior, JT, Cronkite, DA, Papavasiliou, N, and Jeffrey, KL
- Abstract
While interferon (IFN) responses are critical for mammalian antiviral defense, induction of antiviral RNA interference (RNAi) is evident. To date, individual functions of the mammalian RNAi and micro RNA (miRNA) effector proteins Argonautes 1-4 (AGO1-AGO4) during virus infection remain undetermined. AGO2 was recently implicated in mammalian antiviral defense, so we examined antiviral activity of AGO1, AGO3, or AGO4 in IFN-competent immune cells. Only AGO4-deficient cells are hyper-susceptible to virus infection. AGO4 antiviral function is both IFN dependent and IFN independent, since AGO4 promotes IFN but also maintains antiviral capacity following prevention of IFN signaling or production. We identified AGO-loaded virus-derived short interfering RNAs (vsiRNAs), a molecular marker of antiviral RNAi, in macrophages infected with influenza or influenza lacking the IFN and RNAi suppressor NS1, which are uniquely diminished without AGO4. Importantly, AGO4-deficient influenza-infected mice have significantly higher burden and viral titers in vivo. Together, our data assign an essential role for AGO4 in mammalian antiviral defense.
- Published
- 2020
3. A new mouse model for PRPH2 pattern dystrophy exhibits functional compensation prior and subsequent to retinal degeneration.
- Author
-
Cavanaugh BL, Milstein ML, Boucher RC, Tan SX, Hanna MW, Seidel A, Frederiksen R, Saunders TL, Sampath AP, Mitton KP, Zhang DQ, and Goldberg AFX
- Subjects
- Animals, Mice, Retinal Cone Photoreceptor Cells metabolism, Retinal Cone Photoreceptor Cells pathology, Codon, Nonsense, Humans, Retina metabolism, Retina pathology, Retina physiopathology, Mutation, Retinal Rod Photoreceptor Cells metabolism, Retinal Rod Photoreceptor Cells pathology, Retinal Dystrophies genetics, Retinal Dystrophies metabolism, Retinal Dystrophies pathology, Retinal Dystrophies physiopathology, Phenotype, Peripherins genetics, Peripherins metabolism, Disease Models, Animal, Retinal Degeneration genetics, Retinal Degeneration metabolism, Retinal Degeneration physiopathology, Retinal Degeneration pathology, Electroretinography
- Abstract
Mutations in PRPH2 are a relatively common cause of sight-robbing inherited retinal degenerations (IRDs). Peripherin-2 (PRPH2) is a photoreceptor-specific tetraspanin protein that structures the disk rim membranes of rod and cone outer segment (OS) organelles, and is required for OS morphogenesis. PRPH2 is noteworthy for its broad spectrum of disease phenotypes; both inter- and intra-familial heterogeneity have been widely observed and this variability in disease expression and penetrance confounds efforts to understand genotype-phenotype correlations and pathophysiology. Here we report the generation and initial characterization of a gene-edited animal model for PRPH2 disease associated with a nonsense mutation (c.1095:C>A, p.Y285X), which is predicted to truncate the peripherin-2 C-terminal domain. Young (P21) Prph2Y285X/WT mice developed near-normal photoreceptor numbers; however, OS membrane architecture was disrupted, OS protein levels were reduced, and in vivo and ex vivo electroretinography (ERG) analyses found that rod and cone photoreceptor function were each severely reduced. Interestingly, ERG studies also revealed that rod-mediated downstream signaling (b-waves) were functionally compensated in the young animals. This resiliency in retinal function was retained at P90, by which time substantial IRD-related photoreceptor loss had occurred. Altogether, the current studies validate a new mouse model for investigating PRPH2 disease pathophysiology, and demonstrate that rod and cone photoreceptor function and structure are each directly and substantially impaired by the Y285X mutation. They also reveal that Prph2 mutations can induce a functional compensation that resembles homeostatic plasticity, which can stabilize rod-derived signaling, and potentially dampen retinal dysfunction during some PRPH2-associated IRDs., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
4. Neuronal splicing of the unmethylated histone H3K4 reader, PHF21A, prevents excessive synaptogenesis.
- Author
-
Nagai M, Porter RS, Miyasato M, Wang A, Gavilan CM, Hughes ED, Wu MC, Saunders TL, and Iwase S
- Abstract
PHF21A is a histone-binding protein that recognizes unmethylated histone H3K4, the reaction product of LSD1 histone demethylase. PHF21A and LSD1 form a complex, and both undergo neuron-specific microexon splicing. The PHF21A neuronal microexon interferes with nucleosome binding, whereas the LSD1 neuronal microexon weakens H3K4 demethylation activity and can alter the substrate specificity to H3K9 or H4K20. However, the temporal expression patterns of PHF21A and LSD1 splicing isoforms during brain development and their biological roles remain unknown. In this work, we report that neuronal PHF21A isoform expression precedes neuronal LSD1 expression during human neuron differentiation and mouse brain development. The asynchronous splicing events resulted in stepwise deactivation of the LSD1-PHF21A complex in reversing H3K4 methylation. An unbiased proteomics survey revealed that the enzymatically inactive LSD1-PHF21A complex interacts with neuron-specific binding partners, including MYT1-family transcription factors and post-transcriptional mRNA processing proteins such as VIRMA. The interaction with the neuron-specific components, however, did not require the PHF21A microexon, indicating that the neuronal proteomic milieu, rather than the microexon-encoded PHF21A segment, is responsible for neuron-specific complex formation. Finally, by using two Phf21a mutant mouse models, we found that Phf21a neuronal splicing prevents excess synapse formation that otherwise would occur when canonical PHF21A is expressed in neurons. These results suggest that the role of the PHF21A microexon is to dampen LSD1-mediated H3K4 demethylation, thereby containing aberrant synaptogenesis., Competing Interests: Conflict of interest M. C. W. is the CEO of Neurodigitech, LLC. The other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Development of a major histocompatibility complex class II conditional knockout mouse to study cell-specific and time-dependent adaptive immune responses in peripheral nerves.
- Author
-
Ubogu EE, Conner JA, Wang Y, Yadav D, and Saunders TL
- Subjects
- Animals, Mice, Humans, Guillain-Barre Syndrome immunology, Guillain-Barre Syndrome genetics, Guillain-Barre Syndrome pathology, Neuritis, Autoimmune, Experimental immunology, Neuritis, Autoimmune, Experimental pathology, Neuritis, Autoimmune, Experimental genetics, Sciatic Nerve immunology, Sciatic Nerve metabolism, Sciatic Nerve pathology, Peripheral Nerves immunology, Peripheral Nerves metabolism, Peripheral Nerves pathology, Female, Endothelial Cells immunology, Endothelial Cells metabolism, Sural Nerve pathology, Sural Nerve immunology, Male, Time Factors, Adaptive Immunity, Mice, Knockout, Mice, Inbred C57BL, Histocompatibility Antigens Class II metabolism, Histocompatibility Antigens Class II genetics
- Abstract
Introduction/aims: The precise relationship between molecular mimicry and tissue-specific autoimmunity is unknown. Major histocompatibility complex (MHC) class II antigen presenting cell-CD4+ T-cell receptor complex interactions are necessary for adaptive immunity. This study aimed to determine the role of endoneurial endothelial cell MHC class II in autoimmune polyneuropathy., Methods: Cryopreserved Guillain-Barré syndrome (GBS) patient sural nerve biopsies and sciatic nerves from the severe murine experimental autoimmune neuritis (sm-EAN) GBS model were studied. Cultured conditional ready MHC Class II antigen A-alpha chain (H2-Aa) embryonic stem cells were used to generate H2-Aa
flox/+ C57BL/6 mice. Mice were backcrossed and intercrossed to the SJL background to generate H2-Aaflox/flox SJL mice, bred with hemizygous Tamoxifen-inducible von Willebrand factor Cre recombinase (vWF-iCre/+) SJL mice to generate H2-Aaflox/flox ; vWF-iCre/+ mice to study microvascular endothelial cell adaptive immune responses. Sm-EAN was induced in Tamoxifen-treated H2-Aaflox/flox ; vWF-iCre/+, H2-Aaflox/flox ; +/+, H2-Aa+/+ ; vWF-iCre/+ and untreated H2-Aaflox/flox ; vWF-iCre/+ adult female SJL mice. Neurobehavioral, electrophysiological and histopathological assessments were performed at predefined time points., Results: Endoneurial endothelial cell MHC class II expression was observed in normal and inflamed human and mouse peripheral nerves. Tamoxifen-treated H2-Aaflox/flox ; vWF-iCre/+ mice were resistant to sm-EAN despite extensive MHC class II expression in lymphoid and non-lymphoid tissues., Discussion: A conditional MHC class II knockout mouse to study cell- and time-dependent adaptive immune responses in vivo was developed. Initial studies show microvascular endothelial cell MHC class II expression is necessary for peripheral nerve specific autoimmunity, as advocated by human in vitro adaptive immunity and ex vivo transplant rejection studies., (© 2024 The Author(s). Muscle & Nerve published by Wiley Periodicals LLC.)- Published
- 2024
- Full Text
- View/download PDF
6. IKKɛ induces STING non-IFN immune responses via a mechanism analogous to TBK1.
- Author
-
Venkatraman R, Balka KR, Wong W, Sivamani J, Magill Z, Tullett KM, Lane RM, Saunders TL, Tailler M, Crack PJ, Wakim LM, Lahoud MH, Lawlor KE, Kile BT, O'Keeffe M, and De Nardo D
- Abstract
The cGAS-STING pathway responds to cytosolic DNA to elicit host immunity to infection. The activation of stimulator of interferon genes (STING) can trigger a number of critical cellular responses including inflammation, noncanonical autophagy, lipid metabolism, senescence, and cell death. STING-mediated immunity through the production of type I interferons (IFNs) and nuclear factor kappa B (NF-κB)-driven proinflammatory cytokines is primarily driven via the effector protein TBK1. We have previously found that IκBα kinase epsilon (IKKε), a homolog of TBK1, can also facilitate STING-NF-κB responses. Therefore, a thorough understanding of how IKKε participates in STING signaling is essential. Here, we used a combination of genetic and biochemical approaches to provide mechanistic details into how IKKε confers non-IFN (e.g., NF-κB and MAPK) STING responses in macrophages, including in the absence of TBK1. We demonstrate a conserved mechanism of STING binding between TBK1 and IKKε. These findings strengthen our understanding of cGAS-STING signaling and the preservation of host immunity in cases of TBK1-deficiency., Competing Interests: The authors declare no competing interests., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
7. Human SIRT5 variants with reduced stability and activity do not cause neuropathology in mice.
- Author
-
Yuan T, Kumar S, Skinner ME, Victor-Joseph R, Abuaita M, Keijer J, Zhang J, Kunkel TJ, Liu Y, Petrunak EM, Saunders TL, Lieberman AP, Stuckey JA, Neamati N, Al-Murshedi F, Alfadhel M, Spelbrink JN, Rodenburg R, de Boer VCJ, and Lombard DB
- Abstract
SIRT5 is a sirtuin deacylase that removes negatively charged lysine modifications, in the mitochondrial matrix and elsewhere in the cell. In benign cells and mouse models, under basal conditions, the phenotypes of SIRT5 deficiency are quite subtle. Here, we identify two homozygous SIRT5 variants in patients suspected to have mitochondrial disease. Both variants, P114T and L128V, are associated with reduced SIRT5 protein stability and impaired biochemical activity, with no evidence of neomorphic or dominant negative properties. The crystal structure of the P114T enzyme was solved and shows only subtle deviations from wild-type. Via CRISPR-Cas9, we generated a mouse model that recapitulates the human P114T mutation; homozygotes show reduced SIRT5 levels and activity, but no obvious metabolic abnormalities, neuropathology, or other gross phenotypes. We conclude that these human SIRT5 variants most likely represent severe hypomorphs, but are likely not by themselves the primary pathogenic cause of the neuropathology observed in the patients., Competing Interests: The authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
8. Asynchronous microexon splicing of LSD1 and PHF21A during neurodevelopment.
- Author
-
Nagai M, Porter RS, Hughes E, Saunders TL, and Iwase S
- Abstract
LSD1 histone H3K4 demethylase and its binding partner PHF21A, a reader protein for unmethylated H3K4, both undergo neuron-specific microexon splicing. The LSD1 neuronal microexon weakens H3K4 demethylation activity and can alter the substrate specificity to H3K9 or H4K20. Meanwhile, the PHF21A neuronal microexon interferes with nucleosome binding. However, the temporal expression patterns of LSD1 and PHF21A splicing isoforms during brain development remain unknown. In this work, we report that neuronal PHF21A isoform expression precedes neuronal LSD1 isoform expression during human neuron differentiation and mouse brain development. The asynchronous splicing events resulted in stepwise deactivation of the LSD1-PHF21A complex in reversing H3K4 methylation. We further show that the enzymatically inactive LSD1-PHF21A complex interacts with neuron-specific binding partners, including MYT1-family transcription factors and post-transcriptional mRNA processing proteins such as VIRMA. The interaction with the neuron-specific components, however, did not require the PHF21A microexon, indicating that the neuronal proteomic milieu, rather than the microexon-encoded PHF21A segment, is responsible for neuron-specific complex formation. These results indicate that the PHF21A microexon is dispensable for neuron-specific protein-protein interactions, yet the enzymatically inactive LSD1-PHF21A complex might have unique gene-regulatory roles in neurons., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article.
- Published
- 2024
- Full Text
- View/download PDF
9. Exposure of the inner mitochondrial membrane triggers apoptotic mitophagy.
- Author
-
Saunders TL, Windley SP, Gervinskas G, Balka KR, Rowe C, Lane R, Tailler M, Nguyen TN, Ramm G, Lazarou M, De Nardo D, Kile BT, and McArthur K
- Subjects
- Humans, Ubiquitination, Mitochondrial Membranes metabolism, Protein Kinases metabolism, Mitophagy, Ubiquitin-Protein Ligases metabolism
- Abstract
During apoptosis mediated by the intrinsic pathway, BAX/BAK triggers mitochondrial permeabilization and the release of cytochrome-c, followed by a dramatic remodelling of the mitochondrial network that results in mitochondrial herniation and the subsequent release of pro-inflammatory mitochondrial components. Here, we show that mitochondrial herniation and subsequent exposure of the inner mitochondrial membrane (IMM) to the cytoplasm, initiates a unique form of mitophagy to deliver these damaged organelles to lysosomes. IMM-induced mitophagy occurs independently of canonical PINK1/Parkin signalling and is driven by ubiquitination of the IMM. Our data suggest IMM-induced mitophagy is an additional safety mechanism that cells can deploy to contain damaged mitochondria. It may have particular relevance in situations where caspase activation is incomplete or inhibited, and in contexts where PINK1/Parkin-mitophagy is impaired or overwhelmed., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Selective Inhibition of mTORC1 Signaling Supports the Development and Maintenance of Pluripotency.
- Author
-
Kim JK, Villa-Diaz LG, Saunders TL, Saul RP, Timilsina S, Liu F, Mishina Y, and Krebsbach PH
- Subjects
- Humans, Animals, Mice, Sirolimus pharmacology, Phosphorylation, Mechanistic Target of Rapamycin Complex 1, Signal Transduction, Blastocyst
- Abstract
Insight into the molecular mechanisms governing the development and maintenance of pluripotency is important for understanding early development and the use of stem cells in regenerative medicine. We demonstrate the selective inhibition of mTORC1 signaling is important for developing the inner cell mass (ICM) and the self-renewal of human embryonic stem cells. S6K suppressed the expression and function of pluripotency-related transcription factors (PTFs) OCT4, SOX2, and KLF4 through phosphorylation and ubiquitin proteasome-mediated protein degradation, indicating that S6K inhibition is required for pluripotency. PTFs inhibited mTOR signaling. The phosphorylation of S6 was decreased in PTF-positive cells of the ICM in embryos. Activation of mTORC1 signaling blocked ICM formation and the selective inhibition of S6K by rapamycin increased the ICM size in mouse blastocysts. Thus, selective inhibition of mTORC1 signaling supports the development and maintenance of pluripotency., (© The Author(s) 2023. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
11. Development of a C3 Humanized Rat as a New Model for Evaluating Novel C3 Inhibitors.
- Author
-
Chen JY, Zhang L, Yang M, Hughes ED, Freeman ZT, Saunders TL, and Lin F
- Subjects
- Rats, Humans, Animals, Primates, Hemolysis, Complement Activation
- Abstract
Introduction: C3 is central for all complement activation pathways, thus making it an attractive therapeutic target. Many C3-targeted agents are under extensive development with one already approved for clinical use. However, most, if not all, C3 inhibitors are human or nonhuman primate C3-specific, making evaluating their efficacies in vivo before a clinical trial extremely difficult and costly., Methods: We first studied the compatibility of human C3 in the rat complement system, then developed a C3 humanized rat using the CRISPR/Cas9 technology. We thoroughly characterized the resultant human C3 humanized rats and tested the treatment efficacy of an established primate-specific C3 inhibitor in a model of complement-mediated hemolysis in the C3 humanized rats., Results: We found that supplementing human C3 protein into the C3-deficient rat blood restored its complement activity, which was inhibited by rat factor H or compstatin, suggesting that human C3 is compatible to the rat complement system. The newly developed C3 humanized rats appeared healthy and expressed human but not rat C3 without detectable spontaneous C3 activation. More importantly, complement-mediated hemolysis in the C3 humanized rats was also inhibited by compstatin both in vitro and in vivo., Conclusion: The successfully developed C3 humanized rats provided a much-desired rodent model to evaluate novel C3 inhibitors in vivo as potential drugs., (© 2023 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
12. SIRT5 variants from patients with mitochondrial disease are associated with reduced SIRT5 stability and activity, but not with neuropathology.
- Author
-
Yuan T, Kumar S, Skinner M, Victor-Joseph R, Abuaita M, Keijer J, Zhang J, Kunkel TJ, Liu Y, Petrunak EM, Saunders TL, Lieberman AP, Stuckey JA, Neamati N, Al-Murshedi F, Alfadhel M, Spelbrink JN, Rodenburg R, de Boer VCJ, and Lombard DB
- Abstract
SIRT5 is a sirtuin deacylase that represents the major activity responsible for removal of negatively-charged lysine modifications, in the mitochondrial matrix and elsewhere in the cell. In benign cells and mouse models, under basal non-stressed conditions, the phenotypes of SIRT5 deficiency are generally quite subtle. Here, we identify two homozygous SIRT5 variants in human patients suffering from severe mitochondrial disease. Both variants, P114T and L128V, are associated with reduced SIRT5 protein stability and impaired biochemical activity, with no evidence of neomorphic or dominant negative properties. The crystal structure of the P114T enzyme was solved and shows only subtle deviations from wild-type. Via CRISPR-Cas9, we generate a mouse model that recapitulates the human P114T mutation; homozygotes show reduced SIRT5 levels and activity, but no obvious metabolic abnormalities, neuropathology or other gross evidence of severe disease. We conclude that these human SIRT5 variants most likely represent severe hypomorphs, and are likely not the primary pathogenic cause of the neuropathology observed in the patients., Competing Interests: Declaration of Interests: The authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
13. Development of a major histocompatibility complex class II conditional knockout mouse to study cell-specific and time-dependent adaptive immune responses in peripheral nerves.
- Author
-
Ubogu EE, Conner JA, Wang Y, Yadav D, and Saunders TL
- Abstract
Introduction: Major histocompatibility complex (MHC) class II professional antigen presenting cell-naïve CD4+ T cell interactions via the T-cell receptor complex are necessary for adaptive immunity. MHC class II upregulation in multiple cell types occurs in human autoimmune polyneuropathy patient biopsies, necessitating studies to ascertain cellular signaling pathways required for tissue-specific autoimmunity., Methods: Cryopreserved Guillain-Barré syndrome (GBS) patient sural nerve biopsies and sciatic nerves from the severe murine experimental autoimmune neuritis (sm-EAN) GBS model were studied. Cultured conditional ready MHC Class II antigen A-alpha chain (H2-Aa) embryonic stem cells were used to generate H2-Aa
flox/+ C57BL/6 mice. Mice were backcrossed and intercrossed to the SJL background to generate H2-Aaflox/flox SJL mice, bred with hemizygous Tamoxifen-inducible von Willebrand factor Cre recombinase (vWF-iCre/+) SJL mice to generate H2-Aaflox/flox ; vWF-iCre/+ to study microvascular endothelial cell adaptive immune responses. Sm-EAN was induced in adult female SJL Tamoxifen-treated H2-Aaflox/flox ; vWF-iCre/+ mice and H2-Aaflox/flox ; +/+ littermate controls. Neurobehavioral, electrophysiological and histopathological assessments were performed at predefined time points., Results: Endoneurial endothelial cell MHC class II expression was observed in normal and inflamed human and mouse peripheral nerves. Adult female Tamoxifen-treated H2-Aaflox/flox ; vWF-iCre/+ did not develop sm-EAN despite extensive MHC class II expression in lymphoid and non-lymphoid tissues., Discussion: A conditional MHC class II knockout mouse to study cell- and time-dependent adaptive immune responses in vivo is developed. Initial studies show microvascular endothelial cell MHC class II expression is necessary for peripheral nerve specific autoimmunity, as advocated by human in vitro adaptive immunity and ex vivo transplant rejection studies.- Published
- 2023
- Full Text
- View/download PDF
14. Generating endogenous Myh11-driven Cre mice for sex-independent gene deletion in smooth muscle cells.
- Author
-
Zhao Y, Zhao G, Chang Z, Zhu T, Zhao Y, Lu H, Xue C, Saunders TL, Guo Y, Chang L, Chen YE, and Zhang J
- Subjects
- Female, Mice, Male, Animals, Gene Deletion, Mice, Transgenic, Tamoxifen pharmacology, Myocytes, Smooth Muscle
- Abstract
Specific and efficient smooth muscle cell-targeted (SMC-targeted) gene deletion is typically achieved by pairing SMMHC-CreERT2-Tg mice with mice carrying the loxP-flanked gene. However, the transgene, CreERT2, is not controlled by the endogenous Myh11 gene promoter, and the codon-modified iCreERT2 exhibits significant tamoxifen-independent leakage. Furthermore, because the Cre-bearing bacterial artificial chromosome (BAC) is inserted onto the Y chromosome, the SMMHC-CreERT2-Tg mice strain can only exhibit gene deletions in male mice. Additionally, there is a lack of Myh11-driven constitutive Cre mice when tamoxifen usage is a concern. We used CRISPR/Cas9-mediated homologous recombination between a donor vector carrying the CreNLSP2A or CreERT2-P2A sequence and homologous arm surrounding the translation start site of the Myh11 gene to generate Cre-knockin mice. The P2A sequence enables the simultaneous translation of Cre and endogenous proteins. Using reporter mice, we assessed Cre-mediated recombination efficiency, specificity, tamoxifen-dependent controllability, and functionality in both sexes. Both constitutive (Myh11-CreNLSP2A) and inducible (Myh11-CreERT2-P2A) Cre mice demonstrated efficient, SMC-specific, sex-independent Cre recombinase activity without confounding endogenous gene expression. Combined with recently generated BAC transgenic Myh11-CreERT2-RAD mice and the Itga8-CreERT2 mouse models, our models will help expand the research toolbox, facilitating unbiased and comprehensive research in SMCs and SMC-dependent cardiovascular diseases.
- Published
- 2023
- Full Text
- View/download PDF
15. Termination of STING responses is mediated via ESCRT-dependent degradation.
- Author
-
Balka KR, Venkatraman R, Saunders TL, Shoppee A, Pang ES, Magill Z, Homman-Ludiye J, Huang C, Lane RM, York HM, Tan P, Schittenhelm RB, Arumugam S, Kile BT, O'Keeffe M, and De Nardo D
- Subjects
- Mice, Animals, Signal Transduction physiology, Macrophages metabolism, Nucleotidyltransferases metabolism, DNA, Endosomal Sorting Complexes Required for Transport genetics, Immunity, Innate, Membrane Proteins genetics, Membrane Proteins metabolism
- Abstract
cGAS-STING signalling is induced by detection of foreign or mislocalised host double-stranded (ds)DNA within the cytosol. STING acts as the major signalling hub, where it controls production of type I interferons and inflammatory cytokines. Basally, STING resides on the ER membrane. Following activation STING traffics to the Golgi to initiate downstream signalling and subsequently to endolysosomal compartments for degradation and termination of signalling. While STING is known to be degraded within lysosomes, the mechanisms controlling its delivery remain poorly defined. Here we utilised a proteomics-based approach to assess phosphorylation changes in primary murine macrophages following STING activation. This identified numerous phosphorylation events in proteins involved in intracellular and vesicular transport. We utilised high-temporal microscopy to track STING vesicular transport in live macrophages. We subsequently identified that the endosomal complexes required for transport (ESCRT) pathway detects ubiquitinated STING on vesicles, which facilitates the degradation of STING in murine macrophages. Disruption of ESCRT functionality greatly enhanced STING signalling and cytokine production, thus characterising a mechanism controlling effective termination of STING signalling., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
16. Bone marrow endosteal stem cells dictate active osteogenesis and aggressive tumorigenesis.
- Author
-
Matsushita Y, Liu J, Chu AKY, Tsutsumi-Arai C, Nagata M, Arai Y, Ono W, Yamamoto K, Saunders TL, Welch JD, and Ono N
- Subjects
- Adult, Humans, Aged, Bone and Bones, Osteoblasts metabolism, Stem Cells, Carcinogenesis genetics, Carcinogenesis metabolism, Bone Marrow Cells metabolism, Cell Differentiation, Osteogenesis genetics, Bone Marrow metabolism
- Abstract
The bone marrow contains various populations of skeletal stem cells (SSCs) in the stromal compartment, which are important regulators of bone formation. It is well-described that leptin receptor (LepR)
+ perivascular stromal cells provide a major source of bone-forming osteoblasts in adult and aged bone marrow. However, the identity of SSCs in young bone marrow and how they coordinate active bone formation remains unclear. Here we show that bone marrow endosteal SSCs are defined by fibroblast growth factor receptor 3 (Fgfr3) and osteoblast-chondrocyte transitional (OCT) identities with some characteristics of bone osteoblasts and chondrocytes. These Fgfr3-creER-marked endosteal stromal cells contribute to a stem cell fraction in young stages, which is later replaced by Lepr-cre-marked stromal cells in adult stages. Further, Fgfr3+ endosteal stromal cells give rise to aggressive osteosarcoma-like lesions upon loss of p53 tumor suppressor through unregulated self-renewal and aberrant osteogenic fates. Therefore, Fgfr3+ endosteal SSCs are abundant in young bone marrow and provide a robust source of osteoblasts, contributing to both normal and aberrant osteogenesis., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
17. Knockout of murine Lyplal1 confers sex-specific protection against diet-induced obesity.
- Author
-
Vohnoutka RB, Kuppa A, Hegde Y, Chen Y, Pant A, Tohme ME, Choi EK, McCarty SM, Bagchi DP, Du X, Chen Y, Chen VL, Mori H, Bielak LF, Maguire LH, Handelman SK, Sexton JZ, Saunders TL, Halligan BD, and Speliotes EK
- Subjects
- Animals, Female, Humans, Male, Mice, Diet, High-Fat adverse effects, Mice, Inbred C57BL, Mice, Knockout, Triglycerides, Genome-Wide Association Study, Obesity genetics, Obesity metabolism, Lysophospholipase genetics
- Abstract
Human genome-wide association studies found single-nucleotide polymorphisms (SNPs) near LYPLAL1 (Lysophospholipase-like protein 1) that have sex-specific effects on fat distribution and metabolic traits. To determine whether altering LYPLAL1 affects obesity and metabolic disease, we created and characterized a mouse knockout (KO) of Lyplal1. We fed the experimental group of mice a high-fat, high-sucrose (HFHS) diet for 23 weeks, and the controls were fed regular chow diet. Here, we show that CRISPR-Cas9 whole-body Lyplal1 KO mice fed an HFHS diet showed sex-specific differences in weight gain and fat accumulation as compared to chow diet. Female, not male, KO mice weighed less than WT mice, had reduced body fat percentage, had white fat mass, and had adipocyte diameter not accounted for by changes in the metabolic rate. Female, but not male, KO mice had increased serum triglycerides, decreased aspartate, and decreased alanine aminotransferase. Lyplal1 KO mice of both sexes have reduced liver triglycerides and steatosis. These diet-specific effects resemble the effects of SNPs near LYPLAL1 in humans, suggesting that LYPLAL1 has an evolutionary conserved sex-specific effect on adiposity. This murine model can be used to study this novel gene-by-sex-by-diet interaction to elucidate the metabolic effects of LYPLAL1 on human obesity.
- Published
- 2023
- Full Text
- View/download PDF
18. Enamel defects in Acp4 R110C/R110C mice and human ACP4 mutations.
- Author
-
Liang T, Wang SK, Smith C, Zhang H, Hu Y, Seymen F, Koruyucu M, Kasimoglu Y, Kim JW, Zhang C, Saunders TL, Simmer JP, and Hu JC
- Subjects
- Acid Phosphatase metabolism, Ameloblasts metabolism, Amelogenesis, Animals, Histidine metabolism, Humans, Mice, Mutation, Amelogenesis Imperfecta metabolism, Dental Enamel Proteins genetics, Dental Enamel Proteins metabolism
- Abstract
Human ACP4 (OMIM*606362) encodes a transmembrane protein that belongs to histidine acid phosphatase (ACP) family. Recessive mutations in ACP4 cause non-syndromic hypoplastic amelogenesis imperfecta (AI1J, OMIM#617297). While ACP activity has long been detected in developing teeth, its functions during tooth development and the pathogenesis of ACP4-associated AI remain largely unknown. Here, we characterized 2 AI1J families and identified a novel ACP4 disease-causing mutation: c.774_775del, p.Gly260Aspfs*29. To investigate the role of ACP4 during amelogenesis, we generated and characterized Acp4
R110C mice that carry the p.(Arg110Cys) loss-of-function mutation. Mouse Acp4 expression was the strongest at secretory stage ameloblasts, and the protein localized primarily at Tomes' processes. While Acp4 heterozygous (Acp4+/R110C ) mice showed no phenotypes, incisors and molars of homozygous (Acp4R110C/R110C ) mice exhibited a thin layer of aplastic enamel with numerous ectopic mineralized nodules. Acp4R110C/R110C ameloblasts appeared normal initially but underwent pathology at mid-way of secretory stage. Ultrastructurally, sporadic enamel ribbons grew on mineralized dentin but failed to elongate, and aberrant needle-like crystals formed instead. Globs of organic matrix accumulated by the distal membranes of defective Tomes' processes. These results demonstrated a critical role for ACP4 in appositional growth of dental enamel probably by processing and regulating enamel matrix proteins around mineralization front apparatus., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
19. Soluble CD13 induces inflammatory arthritis by activating the bradykinin receptor B1.
- Author
-
Tsou PS, Lu C, Gurrea-Rubio M, Muraoka S, Campbell PL, Wu Q, Model EN, Lind ME, Vichaikul S, Mattichak MN, Brodie WD, Hervoso JL, Ory S, Amarista CI, Pervez R, Junginger L, Ali M, Hodish G, O'Mara MM, Ruth JH, Robida AM, Alt AJ, Zhang C, Urquhart AG, Lawton JN, Chung KC, Maerz T, Saunders TL, Groppi VE, Fox DA, and Amin MA
- Subjects
- Animals, Bradykinin metabolism, Bradykinin pharmacology, Disease Models, Animal, Fibroblasts metabolism, Mice, Receptor, Bradykinin B1 genetics, Receptor, Bradykinin B1 metabolism, Receptors, G-Protein-Coupled metabolism, Synovial Membrane pathology, Arthritis, Rheumatoid pathology, CD13 Antigens metabolism, Synoviocytes metabolism
- Abstract
CD13, an ectoenzyme on myeloid and stromal cells, also circulates as a shed, soluble protein (sCD13) with powerful chemoattractant, angiogenic, and arthritogenic properties, which require engagement of a G protein-coupled receptor (GPCR). Here we identify the GPCR that mediates sCD13 arthritogenic actions as the bradykinin receptor B1 (B1R). Immunofluorescence and immunoblotting verified high expression of B1R in rheumatoid arthritis (RA) synovial tissue and fibroblast-like synoviocytes (FLSs), and demonstrated binding of sCD13 to B1R. Chemotaxis, and phosphorylation of Erk1/2, induced by sCD13, were inhibited by B1R antagonists. In ex vivo RA synovial tissue organ cultures, a B1R antagonist reduced secretion of inflammatory cytokines. Several mouse arthritis models, including serum transfer, antigen-induced, and local innate immune stimulation arthritis models, were attenuated in Cd13-/- and B1R-/- mice and were alleviated by B1R antagonism. These results establish a CD13/B1R axis in the pathogenesis of inflammatory arthritis and identify B1R as a compelling therapeutic target in RA and potentially other inflammatory diseases.
- Published
- 2022
- Full Text
- View/download PDF
20. Human enteric viruses autonomously shape inflammatory bowel disease phenotype through divergent innate immunomodulation.
- Author
-
Adiliaghdam F, Amatullah H, Digumarthi S, Saunders TL, Rahman RU, Wong LP, Sadreyev R, Droit L, Paquette J, Goyette P, Rioux JD, Hodin R, Mihindukulasuriya KA, Handley SA, and Jeffrey KL
- Subjects
- Animals, Humans, Immunomodulation, Inflammation, Mice, Phenotype, Enterovirus, Gastrointestinal Microbiome, Inflammatory Bowel Diseases, Viruses
- Abstract
Altered enteric microorganisms in concert with host genetics shape inflammatory bowel disease (IBD) phenotypes. However, insight is limited to bacteria and fungi. We found that eukaryotic viruses and bacteriophages (collectively, the virome), enriched from non-IBD, noninflamed human colon resections, actively elicited atypical anti-inflammatory innate immune programs. Conversely, ulcerative colitis or Crohn's disease colon resection viromes provoked inflammation, which was successfully dampened by non-IBD viromes. The IBD colon tissue virome was perturbed, including an increase in the enterovirus B species of eukaryotic picornaviruses, not previously detected in fecal virome studies. Mice humanized with non-IBD colon tissue viromes were protected from intestinal inflammation, whereas IBD virome mice exhibited exacerbated inflammation in a nucleic acid sensing-dependent fashion. Furthermore, there were detrimental consequences for IBD patient-derived intestinal epithelial cells bearing loss-of-function mutations within virus sensor MDA5 when exposed to viromes. Our results demonstrate that innate recognition of IBD or non-IBD human viromes autonomously influences intestinal homeostasis and disease phenotypes. Thus, perturbations in the intestinal virome, or an altered ability to sense the virome due to genetic variation, contribute to the induction of IBD. Harnessing the virome may offer therapeutic and biomarker potential.
- Published
- 2022
- Full Text
- View/download PDF
21. Direct cellular reprogramming enables development of viral T antigen-driven Merkel cell carcinoma in mice.
- Author
-
Verhaegen ME, Harms PW, Van Goor JJ, Arche J, Patrick MT, Wilbert D, Zabawa H, Grachtchouk M, Liu CJ, Hu K, Kelly MC, Chen P, Saunders TL, Weidinger S, Syu LJ, Runge JS, Gudjonsson JE, Wong SY, Brownell I, Cieslik M, Udager AM, Chinnaiyan AM, Tsoi LC, and Dlugosz AA
- Subjects
- Animals, Antigens, Viral, Antigens, Viral, Tumor genetics, Antigens, Viral, Tumor metabolism, Cellular Reprogramming, Mice, Carcinoma, Merkel Cell genetics, Carcinoma, Merkel Cell metabolism, Carcinoma, Merkel Cell pathology, Merkel cell polyomavirus genetics, Polyomavirus Infections genetics, Polyomavirus Infections pathology, Skin Neoplasms pathology, Tumor Virus Infections genetics, Tumor Virus Infections pathology
- Abstract
Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer that frequently carries an integrated Merkel cell polyomavirus (MCPyV) genome and expresses viral transforming antigens (TAgs). MCC tumor cells also express signature genes detected in skin-resident, postmitotic Merkel cells, including atonal bHLH transcription factor 1 (ATOH1), which is required for Merkel cell development from epidermal progenitors. We now report the use of in vivo cellular reprogramming, using ATOH1, to drive MCC development from murine epidermis. We generated mice that conditionally expressed MCPyV TAgs and ATOH1 in epidermal cells, yielding microscopic collections of proliferating MCC-like cells arising from hair follicles. Immunostaining of these nascent tumors revealed p53 accumulation and apoptosis, and targeted deletion of transformation related protein 53 (Trp53) led to development of gross skin tumors with classic MCC histology and marker expression. Global transcriptome analysis confirmed the close similarity of mouse and human MCCs, and hierarchical clustering showed conserved upregulation of signature genes. Our data establish that expression of MCPyV TAgs in ATOH1-reprogrammed epidermal cells and their neuroendocrine progeny initiates hair follicle-derived MCC tumorigenesis in adult mice. Moreover, progression to full-blown MCC in this model requires loss of p53, mimicking the functional inhibition of p53 reported in human MCPyV-positive MCCs.
- Published
- 2022
- Full Text
- View/download PDF
22. Slow oscillations persist in pancreatic beta cells lacking phosphofructokinase M.
- Author
-
Marinelli I, Parekh V, Fletcher P, Thompson B, Ren J, Tang X, Saunders TL, Ha J, Sherman A, Bertram R, and Satin LS
- Subjects
- Animals, Calcium metabolism, Glucose metabolism, Insulin metabolism, Insulin Secretion, Mice, Insulin-Secreting Cells metabolism, Islets of Langerhans, Phosphofructokinase-1 genetics, Phosphofructokinase-1 metabolism
- Abstract
Pulsatile insulin secretion by pancreatic beta cells is necessary for tight glucose control in the body. Glycolytic oscillations have been proposed as the mechanism for generating the electrical oscillations underlying pulsatile insulin secretion. The glycolytic enzyme 6-phosphofructokinase-1 (PFK) synthesizes fructose-1,6-bisphosphate (FBP) from fructose-6-phosphate. It has been proposed that the slow electrical and Ca
2+ oscillations (periods of 3-5 min) observed in islets result from allosteric feedback activation of PFKM by FBP. Pancreatic beta cells express three PFK isozymes: PFKL, PFKM, and PFKP. A prior study of mice that were engineered to lack PFKM using a gene-trap strategy to delete Pfkm produced a mosaic reduction in global Pfkm expression, but the islets isolated from the mice still exhibited slow Ca2+ oscillations. However, these islets still expressed residual PFKM protein. Thus, to more fully test the hypothesis that beta cell PFKM is responsible for slow islet oscillations, we made a beta-cell-specific knockout mouse that completely lacked PFKM. While PFKM deletion resulted in subtle metabolic changes in vivo, islets that were isolated from these mice continued to exhibit slow oscillations in electrical activity, beta cell Ca2+ concentrations, and glycolysis, as measured using PKAR, an FBP reporter/biosensor. Furthermore, simulations obtained with a mathematical model of beta cell activity shows that slow oscillations can persist despite PFKM loss provided that one of the other PFK isoforms, such as PFKP, is present, even if its level of expression is unchanged. Thus, while we believe that PFKM may be the main regulator of slow oscillations in wild-type islets, PFKP can provide functional redundancy. Our model also suggests that PFKM likely dominates, in vivo, because it outcompetes PFKP with its higher FBP affinity and lower ATP affinity. We thus propose that isoform redundancy may rescue key physiological processes of the beta cell in the absence of certain critical genes., (Copyright © 2022. Published by Elsevier Inc.)- Published
- 2022
- Full Text
- View/download PDF
23. Angiogenesis depends upon EPHB4-mediated export of collagen IV from vascular endothelial cells.
- Author
-
Chen D, Hughes ED, Saunders TL, Wu J, Vasquez MNH, Makinen T, and King PD
- Subjects
- Animals, Cells, Cultured, DNA Mutational Analysis, Endothelial Cells metabolism, Mice, Mice, Transgenic, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Receptor, EphB4 metabolism, Vascular Malformations metabolism, Vascular Malformations pathology, p120 GTPase Activating Protein deficiency, Collagen Type IV metabolism, DNA genetics, Endothelial Cells pathology, Mutation, Neovascularization, Pathologic genetics, Receptor, EphB4 genetics, Vascular Malformations genetics
- Abstract
Capillary malformation-arteriovenous malformation (CM-AVM) is a blood vascular anomaly caused by inherited loss-of-function mutations in RASA1 or EPHB4 genes, which encode p120 Ras GTPase-activating protein (p120 RasGAP/RASA1) and Ephrin receptor B4 (EPHB4). However, whether RASA1 and EPHB4 function in the same molecular signaling pathway to regulate the blood vasculature is uncertain. Here, we show that induced endothelial cell-specific (EC-specific) disruption of Ephb4 in mice resulted in accumulation of collagen IV in the EC ER, leading to EC apoptotic death and defective developmental, neonatal, and pathological angiogenesis, as reported previously in induced EC-specific RASA1-deficient mice. Moreover, defects in angiogenic responses in EPHB4-deficient mice could be rescued by drugs that inhibit signaling through the Ras pathway and drugs that promote collagen IV export from the ER. However, EPHB4-mutant mice that expressed a form of EPHB4 that is unable to physically engage RASA1 but retains protein tyrosine kinase activity showed normal angiogenic responses. These findings provide strong evidence that RASA1 and EPHB4 function in the same signaling pathway to protect against the development of CM-AVM independent of physical interaction and have important implications for possible means of treatment of this disease.
- Published
- 2022
- Full Text
- View/download PDF
24. The Australasian Cell Death Society (ACDS): celebrating 50 years of Australasian cell death research.
- Author
-
Speir M, Chan AH, Simpson DS, Khan T, Saunders TL, Poon IK, and Atkin-Smith GK
- Subjects
- Cell Death
- Published
- 2022
- Full Text
- View/download PDF
25. SEC23A rescues SEC23B-deficient congenital dyserythropoietic anemia type II.
- Author
-
King R, Lin Z, Balbin-Cuesta G, Myers G, Friedman A, Zhu G, McGee B, Saunders TL, Kurita R, Nakamura Y, Engel JD, Reddy P, and Khoriaty R
- Abstract
Congenital dyserythropoietic anemia type II (CDAII) results from loss-of-function mutations in SEC23B . In contrast to humans, SEC23B-deficient mice deletion do not exhibit CDAII but die perinatally with pancreatic degeneration. Here, we demonstrate that expression of the full SEC23A protein (the SEC23B paralog) from the endogenous regulatory elements of Sec23b completely rescues the SEC23B-deficient mouse phenotype. Consistent with these data, while mice with erythroid-specific deletion of either Sec23a or Sec23b do not exhibit CDAII, we now show that mice with erythroid-specific deletion of all four Sec23 alleles die in mid-embryogenesis with features of CDAII and that mice with deletion of three Sec23 alleles exhibit a milder erythroid defect. To test whether the functional overlap between the SEC23 paralogs is conserved in human erythroid cells, we generated SEC23B-deficient HUDEP-2 cells. Upon differentiation, these cells exhibited features of CDAII, which were rescued by increased expression of SEC23A, suggesting a novel therapeutic strategy for CDAII.
- Published
- 2021
- Full Text
- View/download PDF
26. Mitochondrial complex II in intestinal epithelial cells regulates T cell-mediated immunopathology.
- Author
-
Fujiwara H, Seike K, Brooks MD, Mathew AV, Kovalenko I, Pal A, Lee HJ, Peltier D, Kim S, Liu C, Oravecz-Wilson K, Li L, Sun Y, Byun J, Maeda Y, Wicha MS, Saunders TL, Rehemtulla A, Lyssiotis CA, Pennathur S, and Reddy P
- Subjects
- Animals, Case-Control Studies, Cell Communication, Cells, Cultured, Colitis genetics, Colitis immunology, Colitis pathology, Colon immunology, Colon ultrastructure, Disease Models, Animal, Electron Transport Complex II genetics, Epithelial Cells immunology, Epithelial Cells ultrastructure, Female, Graft vs Host Disease genetics, Graft vs Host Disease immunology, Graft vs Host Disease pathology, Humans, Immunity, Mucosal, Intestinal Mucosa immunology, Intestinal Mucosa ultrastructure, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Mitochondria immunology, Mitochondria ultrastructure, Oxidative Phosphorylation, Succinic Acid metabolism, T-Lymphocytes metabolism, Mice, Colitis enzymology, Colon enzymology, Cytotoxicity, Immunologic, Electron Transport Complex II metabolism, Epithelial Cells enzymology, Graft vs Host Disease enzymology, Intestinal Mucosa enzymology, Mitochondria enzymology, T-Lymphocytes immunology
- Abstract
Intestinal epithelial cell (IEC) damage by T cells contributes to graft-versus-host disease, inflammatory bowel disease and immune checkpoint blockade-mediated colitis. But little is known about the target cell-intrinsic features that affect disease severity. Here we identified disruption of oxidative phosphorylation and an increase in succinate levels in the IECs from several distinct in vivo models of T cell-mediated colitis. Metabolic flux studies, complemented by imaging and protein analyses, identified disruption of IEC-intrinsic succinate dehydrogenase A (SDHA), a component of mitochondrial complex II, in causing these metabolic alterations. The relevance of IEC-intrinsic SDHA in mediating disease severity was confirmed by complementary chemical and genetic experimental approaches and validated in human clinical samples. These data identify a critical role for the alteration of the IEC-specific mitochondrial complex II component SDHA in the regulation of the severity of T cell-mediated intestinal diseases., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
27. Murine SEC24D can substitute functionally for SEC24C during embryonic development.
- Author
-
Adams EJ, Khoriaty R, Kiseleva A, Cleuren ACA, Tomberg K, van der Ent MA, Gergics P, Tang VT, Zhu G, Hoenerhoff MJ, O'Shea KS, Saunders TL, and Ginsburg D
- Subjects
- Animals, Mice, Mice, Transgenic, Embryonic Development, Genetic Complementation Test, Vesicular Transport Proteins biosynthesis, Vesicular Transport Proteins genetics
- Abstract
The COPII component SEC24 mediates the recruitment of transmembrane cargos or cargo adaptors into newly forming COPII vesicles on the ER membrane. Mammalian genomes encode four Sec24 paralogs (Sec24a-d), with two subfamilies based on sequence homology (SEC24A/B and C/D), though little is known about their comparative functions and cargo-specificities. Complete deficiency for Sec24d results in very early embryonic lethality in mice (before the 8 cell stage), with later embryonic lethality (E7.5) observed in Sec24c null mice. To test the potential overlap in function between SEC24C/D, we employed dual recombinase mediated cassette exchange to generate a Sec24c
c-d allele, in which the C-terminal 90% of SEC24C has been replaced by SEC24D coding sequence. In contrast to the embryonic lethality at E7.5 of SEC24C-deficiency, Sec24cc-d/c-d pups survive to term, though dying shortly after birth. Sec24cc-d/c-d pups are smaller in size, but exhibit no other obvious developmental abnormality by pathologic evaluation. These results suggest that tissue-specific and/or stage-specific expression of the Sec24c/d genes rather than differences in cargo export function explain the early embryonic requirements for SEC24C and SEC24D., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
28. Mouse Dspp frameshift model of human dentinogenesis imperfecta.
- Author
-
Liang T, Hu Y, Zhang H, Xu Q, Smith CE, Zhang C, Kim JW, Wang SK, Saunders TL, Lu Y, Hu JC, and Simmer JP
- Subjects
- Animals, Dental Enamel metabolism, Dentin metabolism, Dentinogenesis Imperfecta metabolism, Dentinogenesis Imperfecta physiopathology, Disease Models, Animal, Extracellular Matrix Proteins metabolism, Female, Frameshift Mutation genetics, Humans, Male, Mice, Mice, Transgenic, Phenotype, Phosphoproteins metabolism, Sialoglycoproteins metabolism, Tooth metabolism, Dentinogenesis Imperfecta genetics, Extracellular Matrix Proteins genetics, Phosphoproteins genetics, Sialoglycoproteins genetics
- Abstract
Non-syndromic inherited defects of tooth dentin are caused by two classes of dominant negative/gain-of-function mutations in dentin sialophosphoprotein (DSPP): 5' mutations affecting an N-terminal targeting sequence and 3' mutations that shift translation into the - 1 reading frame. DSPP defects cause an overlapping spectrum of phenotypes classified as dentin dysplasia type II and dentinogenesis imperfecta types II and III. Using CRISPR/Cas9, we generated a Dspp
-1fs mouse model by introducing a FLAG-tag followed by a single nucleotide deletion that translated 493 extraneous amino acids before termination. Developing incisors and/or molars from this mouse and a DsppP19L mouse were characterized by morphological assessment, bSEM, nanohardness testing, histological analysis, in situ hybridization and immunohistochemistry. DsppP19L dentin contained dentinal tubules but grew slowly and was softer and less mineralized than the wild-type. DsppP19L incisor enamel was softer than normal, while molar enamel showed reduced rod/interrod definition. Dspp-1fs dentin formation was analogous to reparative dentin: it lacked dentinal tubules, contained cellular debris, and was significantly softer and thinner than Dspp+/+ and DsppP19L dentin. The Dspp-1fs incisor enamel appeared normal and was comparable to the wild-type in hardness. We conclude that 5' and 3' Dspp mutations cause dental malformations through different pathological mechanisms and can be regarded as distinct disorders., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
29. DSPP dosage affects tooth development and dentin mineralization.
- Author
-
Lim D, Wu KC, Lee A, Saunders TL, and Ritchie HH
- Subjects
- Animals, Chromosomes, Artificial, Bacterial genetics, Collagen Type II, Dentin diagnostic imaging, Dentin pathology, Extracellular Matrix Proteins deficiency, Extracellular Matrix Proteins metabolism, Incisor metabolism, Incisor pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Minerals analysis, Phosphoproteins deficiency, Phosphoproteins metabolism, RNA, Messenger metabolism, Sialoglycoproteins deficiency, Sialoglycoproteins metabolism, Tooth metabolism, X-Ray Microtomography, Dentin metabolism, Extracellular Matrix Proteins genetics, Phosphoproteins genetics, Sialoglycoproteins genetics, Tooth growth & development
- Abstract
Dentin Sialoprotein (DSP) and phosphophoryn (PP) are two most dominant non-collagenous proteins in dentin, which are the cleavage products of the DSPP (dentin sialophosphoprotein) precursor protein. The absence of the DSPP gene in DSPP knock-out (KO) mice results in characteristics that are consistent with dentinogenesis imperfecta type III in humans. Symptoms include thin dentin, bigger pulp chamber with frequent pulp exposure as well as abnormal epithelial-mesenchymal interactions, and the appearance of chondrocyte-like cells in dental pulp. To better understand how DSPP influences tooth development and dentin formation, we used a bacterial artificial chromosome transgene construct (BAC-DSPP) that contained the complete DSPP gene and promoter to generate BAC-DSPP transgenic mice directly in a mouse DSPP KO background. Two BAC-DSPP transgenic mouse strains were generated and characterized. DSPP mRNA expression in BAC-DSPP Strain A incisors was similar to that from wild-type (wt) mice. DSPP mRNA expression in BAC-DSPP Strain B animals was only 10% that of wt mice. PP protein content in Strain A incisors was 25% of that found in wt mice, which was sufficient to completely rescue the DSPP KO defect in mineral density, since microCT dentin mineral density analysis in 21-day postnatal animal molars showed essentially identical mineral density in both strain A and wt mice. Strain B mouse incisors, with 5% PP expression, only partially rescued the DSPP KO defect in mineral density, as microCT scans of 21-day postnatal animal molars indicated a reduced dentin mineral density compared to wt mice, though the mineral density was still increased over that of DSPP KO. Furthermore, our findings showed that DSPP dosage in Strain A was sufficient to rescue the DSPP KO defect in terms of epithelial-mesenchymal interactions, odontoblast lineage maintenance, along with normal dentin thickness and normal mineral density while DSPP gene dosage in Strain B only partially rescued the aforementioned DSPP KO defect., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
30. Generation of Murine Bone Marrow and Fetal Liver Chimeras.
- Author
-
Chappaz S, Saunders TL, and Kile BT
- Subjects
- Animals, Hematopoietic Stem Cells, Liver, Mice, Radiation Chimera, Bone Marrow, Chimera
- Abstract
The generation of radiation chimeras allows researchers to substitute the hematopoietic system of a mouse with that of one or more donors. A suspension of hematopoietic stem cells (HSCs) is prepared from the bone marrow (BM) or the fetal liver (FL) of a donor mouse and adoptively transferred into an irradiated recipient. Within days, the donor's HSCs will engraft, and their progeny will quickly replace the blood cells of the recipient. This simple tool, together with the large availability of genetically modified mouse lines, can be harnessed to manipulate and study various aspects of blood cell biology in vivo. We present here protocols to generate three types of radiation chimera: (1) BM chimeras, which can assist in determining whether the origin of a genetically based phenotype is the hematopoietic or radio-resistant compartment and which are also conducive for studying the ecology of blood cells and for manipulating the environment hematopoietic cells live; (2) FL chimeras, which allow the study of hematopoietic systems from animals that carry genetic modifications incompatible with postnatal life; and (3) mixed BM chimeras, in which the hematopoietic system comprises blood cells of two different genotypes. Mixed BM chimeras can be used to identify genes that affect hematopoietic cell fitness and to establish whether secreted factors mediate a phenotype of interest. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Generation of bone marrow chimera Basic Protocol 2: Generation of fetal liver chimera Basic Protocol 3: Generation of mixed bone marrow chimera Support Protocol 1: Isolation of bone marrow cells Support Protocol 2: Cell counting by flow cytometry Support Protocol 3: Assessment of chimerism., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
31. Designing and generating a mouse model: frequently asked questions.
- Author
-
Gurumurthy CB, Saunders TL, and Ohtsuka M
- Abstract
Genetically engineered mouse (GEM) models are commonly used in biomedical research. Generating GEMs involve complex set of experimental procedures requiring sophisticated equipment and highly skilled technical staff. Because of these reasons, most research institutes set up centralized core facilities where custom GEMs are created for research groups. Researchers, on the other hand, when they begin thinking about generating GEMs for their research, several questions arise in their minds. For example, what type of model(s) would be best useful for my research, how do I design them, what are the latest technologies and tools available for developing my model(s), and finally how to breed GEMs in my research. As there are several considerations and options in mouse designs, and as it is an expensive and time-consuming endeavor, careful planning upfront can ensure the highest chance of success. In this article, we provide brief answers to several frequently asked questions that arise when researchers begin thinking about generating mouse model(s) for their work.
- Published
- 2021
- Full Text
- View/download PDF
32. Odontogenesis-associated phosphoprotein truncation blocks ameloblast transition into maturation in Odaph C41*/C41* mice.
- Author
-
Liang T, Hu Y, Kawasaki K, Zhang H, Zhang C, Saunders TL, Simmer JP, and Hu JC
- Subjects
- Amelogenesis Imperfecta genetics, Amelogenesis Imperfecta pathology, Animals, Dental Enamel growth & development, Dental Enamel ultrastructure, Extracellular Matrix Proteins chemistry, Extracellular Matrix Proteins genetics, Gene Knock-In Techniques, In Situ Hybridization, Incisor anatomy & histology, Mice, Molar anatomy & histology, Odontogenesis, Phosphoproteins chemistry, Phosphoproteins genetics, Ameloblasts physiology, Amelogenesis, Extracellular Matrix Proteins metabolism, Phosphoproteins metabolism
- Abstract
Mutations of Odontogenesis-Associated Phosphoprotein (ODAPH, OMIM *614829) cause autosomal recessive amelogenesis imperfecta, however, the function of ODAPH during amelogenesis is unknown. Here we characterized normal Odaph expression by in situ hybridization, generated Odaph truncation mice using CRISPR/Cas9 to replace the TGC codon encoding Cys41 into a TGA translation termination codon, and characterized and compared molar and incisor tooth formation in Odaph
+/+ , Odaph+/C41* , and OdaphC41*/C41* mice. We also searched genomes to determine when Odaph first appeared phylogenetically. We determined that tooth development in Odaph+/+ and Odaph+/C41* mice was indistinguishable in all respects, so the condition in mice is inherited in a recessive pattern, as it is in humans. Odaph is specifically expressed by ameloblasts starting with the onset of post-secretory transition and continues until mid-maturation. Based upon histological and ultrastructural analyses, we determined that the secretory stage of amelogenesis is not affected in OdaphC41*/C41* mice. The enamel layer achieves a normal shape and contour, normal thickness, and normal rod decussation. The fundamental problem in OdaphC41*/C41* mice starts during post-secretory transition, which fails to generate maturation stage ameloblasts. At the onset of what should be enamel maturation, a cyst forms that separates flattened ameloblasts from the enamel surface. The maturation stage fails completely.- Published
- 2021
- Full Text
- View/download PDF
33. In Vivo CRISPR/Cas9-Based Targeted Disruption and Knockin of a Long Noncoding RNA.
- Author
-
Cheng X, Peters ST, Pruett-Miller SM, Saunders TL, and Joe B
- Subjects
- Animals, CRISPR-Cas Systems, Embryonic Development, Gene Knock-In Techniques, Rats, Rats, Inbred Dahl, Sequence Deletion, Gene Editing methods, RNA, Long Noncoding genetics
- Abstract
The CRISPR/Cas9 system has been widely used as an efficient genome-editing tool for studying physiological functions of long noncoding RNAs (lncRNAs). In this chapter, we describe the experimental procedures for using the CRISPR/Cas9 system to genetically modify a long noncoding RNA in vivo through the targeted disruption and knockin approaches.
- Published
- 2021
- Full Text
- View/download PDF
34. Histone Acetyltransferase MOF Blocks Acquisition of Quiescence in Ground-State ESCs through Activating Fatty Acid Oxidation.
- Author
-
Khoa LTP, Tsan YC, Mao F, Kremer DM, Sajjakulnukit P, Zhang L, Zhou B, Tong X, Bhanu NV, Choudhary C, Garcia BA, Yin L, Smith GD, Saunders TL, Bielas SL, Lyssiotis CA, and Dou Y
- Subjects
- Cell Differentiation, Cell Division, Fatty Acids, Humans, Embryonic Stem Cells, Histone Acetyltransferases genetics
- Abstract
Self-renewing embryonic stem cells (ESCs) respond to environmental cues by exiting pluripotency or entering a quiescent state. The molecular basis underlying this fate choice remains unclear. Here, we show that histone acetyltransferase MOF plays a critical role in this process through directly activating fatty acid oxidation (FAO) in the ground-state ESCs. We further show that the ground-state ESCs particularly rely on elevated FAO for oxidative phosphorylation (OXPHOS) and energy production. Mof deletion or FAO inhibition induces bona fide quiescent ground-state ESCs with an intact core pluripotency network and transcriptome signatures akin to the diapaused epiblasts in vivo. Mechanistically, MOF/FAO inhibition acts through reducing mitochondrial respiration (i.e., OXPHOS), which in turn triggers reversible pluripotent quiescence specifically in the ground-state ESCs. The inhibition of FAO/OXPHOS also induces quiescence in naive human ESCs. Our study suggests a general function of the MOF/FAO/OXPHOS axis in regulating cell fate determination in stem cells., Competing Interests: Declaration of Interests The authors declare no competing interests, (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
35. TBK1 and IKKε Act Redundantly to Mediate STING-Induced NF-κB Responses in Myeloid Cells.
- Author
-
Balka KR, Louis C, Saunders TL, Smith AM, Calleja DJ, D'Silva DB, Moghaddas F, Tailler M, Lawlor KE, Zhan Y, Burns CJ, Wicks IP, Miner JJ, Kile BT, Masters SL, and De Nardo D
- Subjects
- Animals, Female, HEK293 Cells, Humans, I-kappa B Kinase physiology, Immunity, Innate, Interferon Regulatory Factor-3 metabolism, Interferon-beta metabolism, Male, Membrane Proteins metabolism, Membrane Proteins physiology, Mice, Mice, Inbred C57BL, Myeloid Cells metabolism, NF-kappa B metabolism, Nucleotides, Cyclic metabolism, Phosphorylation, Protein Serine-Threonine Kinases physiology, Signal Transduction immunology, I-kappa B Kinase metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Stimulator of Interferon Genes (STING) is a critical component of host innate immune defense but can contribute to chronic autoimmune or autoinflammatory disease. Once activated, the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS)-STING pathway induces both type I interferon (IFN) expression and nuclear factor-κB (NF-κB)-mediated cytokine production. Currently, these two signaling arms are thought to be mediated by a single upstream kinase, TANK-binding kinase 1 (TBK1). Here, using genetic and pharmacological approaches, we show that TBK1 alone is dispensable for STING-induced NF-κB responses in human and mouse immune cells, as well as in vivo. We further demonstrate that TBK1 acts redundantly with IκB kinase ε (IKKε) to drive NF-κB upon STING activation. Interestingly, we show that activation of IFN regulatory factor 3 (IRF3) is highly dependent on TBK1 kinase activity, whereas NF-κB is significantly less sensitive to TBK1/IKKε kinase inhibition. Our work redefines signaling events downstream of cGAS-STING. Our findings further suggest that cGAS-STING will need to be targeted directly to effectively ameliorate the inflammation underpinning disorders associated with STING hyperactivity., Competing Interests: Declaration of Interests S.L.M. receives funding from GlaxoSmithKline., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
36. Principles of Genetic Engineering.
- Author
-
Lanigan TM, Kopera HC, and Saunders TL
- Subjects
- Animals, CRISPR-Cas Systems, Gene Targeting methods, Gene Transfer Techniques, Genetic Engineering standards, Genetic Engineering trends, Humans, Genetic Engineering methods
- Abstract
Genetic engineering is the use of molecular biology technology to modify DNA sequence(s) in genomes, using a variety of approaches. For example, homologous recombination can be used to target specific sequences in mouse embryonic stem (ES) cell genomes or other cultured cells, but it is cumbersome, poorly efficient, and relies on drug positive/negative selection in cell culture for success. Other routinely applied methods include random integration of DNA after direct transfection (microinjection), transposon-mediated DNA insertion, or DNA insertion mediated by viral vectors for the production of transgenic mice and rats. Random integration of DNA occurs more frequently than homologous recombination, but has numerous drawbacks, despite its efficiency. The most elegant and effective method is technology based on guided endonucleases, because these can target specific DNA sequences. Since the advent of clustered regularly interspaced short palindromic repeats or CRISPR/Cas9 technology, endonuclease-mediated gene targeting has become the most widely applied method to engineer genomes, supplanting the use of zinc finger nucleases, transcription activator-like effector nucleases, and meganucleases. Future improvements in CRISPR/Cas9 gene editing may be achieved by increasing the efficiency of homology-directed repair. Here, we describe principles of genetic engineering and detail: (1) how common elements of current technologies include the need for a chromosome break to occur, (2) the use of specific and sensitive genotyping assays to detect altered genomes, and (3) delivery modalities that impact characterization of gene modifications. In summary, while some principles of genetic engineering remain steadfast, others change as technologies are ever-evolving and continue to revolutionize research in many fields.
- Published
- 2020
- Full Text
- View/download PDF
37. In vivo glucoregulation and tissue-specific glucose uptake in female Akt substrate 160 kDa knockout rats.
- Author
-
Zheng X, Arias EB, Qi NR, Saunders TL, and Cartee GD
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Disease Models, Animal, Female, GTPase-Activating Proteins genetics, Glucose Clamp Technique, Glucose Tolerance Test, Glucose Transport Proteins, Facilitative metabolism, Humans, Liver metabolism, Physical Conditioning, Animal, Rats, Rats, Transgenic, Rats, Wistar, Signal Transduction, GTPase-Activating Proteins deficiency, Gluconeogenesis genetics, Glucose metabolism, Insulin Resistance genetics, Muscle, Skeletal metabolism
- Abstract
The Rab GTPase activating protein known as Akt substrate of 160 kDa (AS160 or TBC1D4) regulates insulin-stimulated glucose uptake in skeletal muscle, the heart, and white adipose tissue (WAT). A novel rat AS160-knockout (AS160-KO) was created with CRISPR/Cas9 technology. Because female AS160-KO versus wild type (WT) rats had not been previously evaluated, the primary objective of this study was to compare female AS160-KO rats with WT controls for multiple, important metabolism-related endpoints. Body mass and composition, physical activity, and energy expenditure were not different between genotypes. AS160-KO versus WT rats were glucose intolerant based on an oral glucose tolerance test (P<0.001) and insulin resistant based on a hyperinsulinemic-euglycemic clamp (HEC; P<0.001). Tissue glucose uptake during the HEC of female AS160-KO versus WT rats was: 1) significantly lower in epitrochlearis (P<0.05) and extensor digitorum longus (EDL; P<0.01) muscles of AS160-KO compared to WT rats; 2) not different in soleus, gastrocnemius or WAT; and 3) ~3-fold greater in the heart (P<0.05). GLUT4 protein content was reduced in AS160-KO versus WT rats in the epitrochlearis (P<0.05), EDL (P<0.05), gastrocnemius (P<0.05), soleus (P<0.05), WAT (P<0.05), and the heart (P<0.005). Insulin-stimulated glucose uptake by isolated epitrochlearis and soleus muscles was lower (P<0.001) in AS160-KO versus WT rats. Akt phosphorylation of insulin-stimulated tissues was not different between the genotypes. A secondary objective was to probe processes that might account for the genotype-related increase in myocardial glucose uptake, including glucose transporter protein abundance (GLUT1, GLUT4, GLUT8, SGLT1), hexokinase II protein abundance, and stimulation of the AMP-activated protein kinase (AMPK) pathway. None of these parameters differed between genotypes. Metabolic phenotyping in the current study revealed AS160 deficiency produced a profound glucoregulatory phenotype in female AS160-KO rats that was strikingly similar to the results previously reported in male AS160-KO rats., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
38. A Requirement for Argonaute 4 in Mammalian Antiviral Defense.
- Author
-
Adiliaghdam F, Basavappa M, Saunders TL, Harjanto D, Prior JT, Cronkite DA, Papavasiliou N, and Jeffrey KL
- Subjects
- Animals, Antiviral Agents pharmacology, Argonaute Proteins pharmacology, Mice, Antiviral Agents therapeutic use, Argonaute Proteins therapeutic use, RNA Interference immunology
- Abstract
While interferon (IFN) responses are critical for mammalian antiviral defense, induction of antiviral RNA interference (RNAi) is evident. To date, individual functions of the mammalian RNAi and micro RNA (miRNA) effector proteins Argonautes 1-4 (AGO1-AGO4) during virus infection remain undetermined. AGO2 was recently implicated in mammalian antiviral defense, so we examined antiviral activity of AGO1, AGO3, or AGO4 in IFN-competent immune cells. Only AGO4-deficient cells are hyper-susceptible to virus infection. AGO4 antiviral function is both IFN dependent and IFN independent, since AGO4 promotes IFN but also maintains antiviral capacity following prevention of IFN signaling or production. We identified AGO-loaded virus-derived short interfering RNAs (vsiRNAs), a molecular marker of antiviral RNAi, in macrophages infected with influenza or influenza lacking the IFN and RNAi suppressor NS1, which are uniquely diminished without AGO4. Importantly, AGO4-deficient influenza-infected mice have significantly higher burden and viral titers in vivo. Together, our data assign an essential role for AGO4 in mammalian antiviral defense., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Murine Surf4 is essential for early embryonic development.
- Author
-
Emmer BT, Lascuna PJ, Tang VT, Kotnik EN, Saunders TL, Khoriaty R, and Ginsburg D
- Subjects
- Alleles, Animals, Apolipoproteins B blood, Apolipoproteins B metabolism, CRISPR-Cas Systems genetics, Cholesterol blood, Gene Editing, Heterozygote, Membrane Proteins metabolism, Mice, Mice, Knockout, Proprotein Convertase 9 blood, Proprotein Convertase 9 metabolism, Embryonic Development, Membrane Proteins genetics
- Abstract
Newly synthesized proteins co-translationally inserted into the endoplasmic reticulum (ER) lumen may be recruited into anterograde transport vesicles by their association with specific cargo receptors. We recently identified a role for the cargo receptor SURF4 in facilitating the secretion of PCSK9 in cultured cells. To examine the function of SURF4 in vivo, we used CRISPR/Cas9-mediated gene editing to generate mice with germline loss-of-function mutations in Surf4. Heterozygous Surf4+/- mice exhibit grossly normal appearance, behavior, body weight, fecundity, and organ development, with no significant alterations in circulating plasma levels of PCSK9, apolipoprotein B, or total cholesterol, and a detectable accumulation of intrahepatic apoliprotein B. Homozygous Surf4-/- mice exhibit embryonic lethality, with complete loss of all Surf4-/- offspring between embryonic days 3.5 and 9.5. In contrast to the milder murine phenotypes associated with deficiency of known SURF4 cargoes, the embryonic lethality of Surf4-/- mice implies the existence of additional SURF4 cargoes or functions that are essential for murine early embryonic development., Competing Interests: DG is a Howard Hughes Medical Institute investigator. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2020
- Full Text
- View/download PDF
40. The History of Transgenesis.
- Author
-
Saunders TL
- Subjects
- Animals, Embryonic Stem Cells cytology, History, 20th Century, History, 21st Century, Mice, Microinjections, Zygote growth & development, Embryo Transfer methods, Gene Transfer Techniques history, Mice, Transgenic genetics, Transgenes genetics
- Abstract
A transgenic mouse carries within its genome an artificial DNA construct (transgene) that is deliberately introduced by an experimentalist. These animals are widely used to understand gene function and protein function. When addressing the history of transgenic mouse technology, it is apparent that a number of basic science research areas laid the groundwork for success. These include reproductive science, genetics and molecular biology, and micromanipulation and microscopy equipment. From reproductive physiology came applications on how to optimize mouse breeding, how to superovulate mice to produce zygotes for DNA microinjection or preimplantation embryos for combination with embryonic stem (ES) cells, and how to return zygotes and embryos to a pseudopregnant surrogate dam for gestation and birth. From developmental biology, it was learned how to micromanipulate embryos for morula aggregation and blastocyst microinjection and how to establish germline competent ES cells. From genetics came the foundational principles governing the inheritance of genes, the interactions of gene products, and an understanding of the phenotypic consequences of genetic mutations. From molecular biology came a panoply of tools and reagents that are used to clone DNA transgenes, to detect the presence of transgenes, to assess gene expression by measuring transcription, and to detect proteins in cells and tissues. Technical advances in light microscopes, micromanipulators, micropipette pullers, and ancillary equipment made it possible for experimentalists to insert thin glass needles into zygotes or embryos under controlled conditions to inject DNA solutions or ES cells. To fully discuss the breadth of contributions of these numerous scientific disciplines to a comprehensive history of transgenic science is beyond the scope of this work. Examples will be used to illustrate scientific developments central to the foundation of transgenic technology and that are in use today.
- Published
- 2020
- Full Text
- View/download PDF
41. Thioredoxin and aging: What have we learned from the survival studies?
- Author
-
Roman MG, Flores LC, Cunningham GM, Cheng C, Allen C, Hubbard GB, Bai Y, Saunders TL, and Ikeno Y
- Abstract
Our laboratory has conducted the first systematic survival studies to examine the biological effects of the antioxidant protein thioredoxin (Trx) on aging and age-related pathology. Our studies with C57BL/6 mice overexpressing Trx1 [Tg(act-TRX1)
+/0 and Tg(TXN)+/0 ) demonstrated a slight extension in early lifespan compared to wild-type (WT) mice; however, no significant effects were observed in the later part of life. Overexpression of Trx2 in male C57BL/6 mice [Tg(TXN2)+/0 ] demonstrated a slightly extended lifespan compared to WT mice. The pathology results from two lines of Trx1 transgenic mice showed a slightly higher incidence of age-related neoplastic diseases compared to WT mice, and a slight increase in the severity of lymphoma, a major neoplastic disease, was observed in Trx2 transgenic mice. Together these studies indicate that Trx overexpression in one compartment of the cell (cytosol or mitochondria alone) has marginal beneficial effects on lifespan. On the other hand, down-regulation of Trx in either the cytosol (Trx1KO) or mitochondria (Trx2KO) showed no significant changes in lifespan compared to WT mice, despite several changes in pathophysiology of these knockout mice. When we examined the synergetic effects of overexpressing Trx1 and Trx2, TXNTg x TXN2Tg mice showed a significantly shorter lifespan with accelerated cancer development compared to WT mice. These results suggest that synergetic effects of Trx overexpression in both the cytosol and mitochondria on aging are deleterious and the development of age-related cancer is accelerated. On the other hand, we have recently found that down-regulation of Trx in both the cytosol and mitochondria in Trx1KO x Trx2KO mice has beneficial effects on aging. The results generated from our lab along with our ongoing study using Trx1KO x Trx2KO mice could elucidate the key pathways (i.e., apoptosis and autophagy) that prevent accumulation of damaged cells and genomic instability leading to reduced cancer formation., Competing Interests: Conflict of interest: The authors declare that they have no conflict of interest.- Published
- 2020
- Full Text
- View/download PDF
42. Thioredoxin overexpression in mitochondria showed minimum effects on aging and age-related diseases in male C57BL/6 mice.
- Author
-
Roman MG, Flores LC, Cunningham GM, Cheng C, Dube S, Allen C, Van Remmen H, Bai Y, Hubbard GB, Saunders TL, and Ikeno Y
- Abstract
Objective: In this study, the effects of overexpression of thioredoxin 2 (Trx2) on aging and age-related diseases were examined using Trx2 transgenic mice [Tg(TXN2]
+/0 ]. Because our previous studies demonstrated that thioredoxin (Trx) overexpression in the cytosol (Trx1) did not extend maximum lifespan, this study was conducted to test if increased Trx2 expression in mitochondria shows beneficial effects on aging and age-related pathology., Methods: Trx2 transgenic mice were generated using a fragment of the human genome containing the TXN2 gene. Effects of Trx2 overexpression on survival, age-related pathology, oxidative stress, and redox-sensitive signaling pathways were examined in male Tg(TXN2)+/0 mice., Results: Trx2 levels were significantly higher (approximately 1.6- to 5-fold) in all of the tissues we examined in Tg(TXN2)+/0 mice compared to wild-type (WT) littermates, and the expression levels were maintained during aging (up to 22-24 months old). Trx2 overexpression did not alter the levels of Trx1, glutaredoxin, glutathione, or other major antioxidant enzymes. Overexpression of Trx2 was associated with reduced reactive oxygen species (ROS) production from mitochondria and lower isoprostane levels compared to WT mice. When we conducted the survival study, male Tg(TXN2)+/0 mice showed a slight extension (approximately 8-9%] of mean, median, and 10th percentile lifespans; however, the survival curve was not significantly different from WT mice. Cross-sectional pathological analysis (22-24 months old) showed that Tg(TXN2)+/0 mice had a slightly higher severity of lymphoma; however, tumor burden, disease burden, and severity of glomerulonephritis and inflammation were similar to WT mice. Trx2 overexpression was also associated with higher c-Jun and c-Fos levels; however, mTOR activity and levels of NFκB p65 and p50 were similar to WT littermates., Conclusions: Our findings suggest that the increased levels of Trx2 in mitochondria over the lifespan in Tg(TXN2)+/0 mice showed a slight life-extending effect, reduced ROS production from mitochondria and oxidative damage to lipids, but showed no significant effects on aging and age-related diseases., Competing Interests: Conflict of Interest: The authors declare that they have no conflict of interest.- Published
- 2020
- Full Text
- View/download PDF
43. MLL1 Inhibition and Vitamin D Signaling Cooperate to Facilitate the Expanded Pluripotency State.
- Author
-
Zhang H, Khoa LTP, Mao F, Xu H, Zhou B, Han Y, O'Leary M, Nusrat A, Wang L, Saunders TL, and Dou Y
- Subjects
- Cell Differentiation, Humans, Signal Transduction, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Myeloid-Lymphoid Leukemia Protein antagonists & inhibitors, Pluripotent Stem Cells metabolism, Vitamin D metabolism
- Abstract
Dynamic establishment of histone modifications in early development coincides with programed cell fate restriction and loss of totipotency beyond the early blastocyst stage. Causal function of histone-modifying enzymes in this process remains to be defined. Here we show that inhibiting histone methyltransferase MLL1 reprograms naive embryonic stem cells (ESCs) to expanded pluripotent stem cells (EPSCs), with differentiation potential toward both embryonic and extraembryonic lineages in vitro and in vivo. MLL1 inhibition or deletion upregulates gene signatures of early blastomere development. The function of MLL1 in restricting induction of EPSCs is mediated partly by Gc, which regulates cellular response to vitamin D signaling. Combined treatment of MLL1 inhibitor and 1α,25-dihydroxyvitamin D
3 (1,25-(OH)2 D3 ) cooperatively enhanced functionality of EPSCs, triggering an extended 2C-like state in vitro and robust totipotent-like property in vivo. Our study sheds light on interplay between epigenetics and vitamin D pathway in cell fate determination., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
44. Knock-In Rat Lines with Cre Recombinase at the Dopamine D1 and Adenosine 2a Receptor Loci.
- Author
-
Pettibone JR, Yu JY, Derman RC, Faust TW, Hughes ED, Filipiak WE, Saunders TL, Ferrario CR, and Berke JD
- Subjects
- Animals, Female, Gene Knock-In Techniques methods, Integrases biosynthesis, Male, Rats, Rats, Long-Evans, Rats, Transgenic, Receptor, Adenosine A2A biosynthesis, Receptors, Dopamine D1 biosynthesis, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Integrases genetics, Receptor, Adenosine A2A genetics, Receptors, Dopamine D1 genetics
- Abstract
Genetically modified mice have become standard tools in neuroscience research. Our understanding of the basal ganglia in particular has been greatly assisted by BAC mutants with selective transgene expression in striatal neurons forming the direct or indirect pathways. However, for more sophisticated behavioral tasks and larger intracranial implants, rat models are preferred. Furthermore, BAC lines can show variable expression patterns depending upon genomic insertion site. We therefore used CRISPR/Cas9 to generate two novel knock-in rat lines specifically encoding Cre recombinase immediately after the dopamine D1 receptor ( Drd1a ) or adenosine 2a receptor ( Adora2a ) loci. Here, we validate these lines using in situ hybridization and viral vector mediated transfection to demonstrate selective, functional Cre expression in the striatal direct and indirect pathways, respectively. We used whole-genome sequencing to confirm the lack of off-target effects and established that both rat lines have normal locomotor activity and learning in simple instrumental and Pavlovian tasks. We expect these new D1-Cre and A2a-Cre rat lines will be widely used to study both normal brain functions and neurological and psychiatric pathophysiology., (Copyright © 2019 Pettibone et al.)
- Published
- 2019
- Full Text
- View/download PDF
45. Absence of complement component 3 does not prevent classical pathway-mediated hemolysis.
- Author
-
Zhang L, Dai Y, Huang P, Saunders TL, Fox DA, Xu J, and Lin F
- Subjects
- Animals, Complement C3 antagonists & inhibitors, Complement C3 therapeutic use, Complement C5 drug effects, Female, Hemolysis drug effects, Humans, Male, Models, Animal, Rats, Complement C3 metabolism, Complement C3-C5 Convertases metabolism, Complement C5 metabolism, Hemolysis immunology
- Abstract
Complement component 3 (C3) is emerging as a potential therapeutic target. We studied complement-mediated hemolysis using normal and C3-depleted human sera, wild-type (WT) and C3-deficient rat sera, and WT and C3 knockout rat models. In all of the in vitro and in vivo experiments, we found that the loss of C3 did not prevent classical pathway-mediated hemolysis, but it did almost abolish alternative pathway-mediated hemolysis. Experiments using preassembled classical pathway C3 convertases confirmed that C4b2a directly activated complement component 5 (C5), leading to membrane attack complex formation and hemolysis. Our results suggest that targeting C3 should effectively inhibit hemolysis and tissue damage mediated by the alternative pathway of complement activation, but this approach might have limited efficacy in treating classical pathway-mediated pathological conditions., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
46. An upstream enhancer regulates Gpihbp1 expression in a tissue-specific manner.
- Author
-
Allan CM, Heizer PJ, Tu Y, Sandoval NP, Jung RS, Morales JE, Sajti E, Troutman TD, Saunders TL, Cusanovich DA, Beigneux AP, Romanoski CE, Fong LG, and Young SG
- Subjects
- Animals, CRISPR-Cas Systems genetics, Chromatin genetics, Heart, Humans, Mice, Mice, Inbred Strains, Receptors, Lipoprotein analysis, Receptors, Lipoprotein metabolism, Sequence Analysis, DNA, Triglycerides blood, Triglycerides metabolism, Adipose Tissue, Brown metabolism, Lipoprotein Lipase metabolism, Receptors, Lipoprotein genetics
- Abstract
Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), the protein that shuttles LPL to the capillary lumen, is essential for plasma triglyceride metabolism. When GPIHBP1 is absent, LPL remains stranded within the interstitial spaces and plasma triglyceride hydrolysis is impaired, resulting in severe hypertriglyceridemia. While the functions of GPIHBP1 in intravascular lipolysis are reasonably well understood, no one has yet identified DNA sequences regulating GPIHBP1 expression. In the current studies, we identified an enhancer element located ∼3.6 kb upstream from exon 1 of mouse Gpihbp1. To examine the importance of the enhancer, we used CRISPR/Cas9 genome editing to create mice lacking the enhancer ( Gpihbp1
Enh/Enh ). Removing the enhancer reduced Gpihbp1 expression by >90% in the liver and by ∼50% in heart and brown adipose tissue. The reduced expression of GPIHBP1 was insufficient to prevent LPL from reaching the capillary lumen, and it did not lead to hypertriglyceridemia-even when mice were fed a high-fat diet. Compound heterozygotes ( Gpihbp1Enh/- mice) displayed further reductions in Gpihbp1 expression and exhibited partial mislocalization of LPL (increased amounts of LPL within the interstitial spaces of the heart), but the plasma triglyceride levels were not perturbed. The enhancer element that we identified represents the first insight into DNA sequences controlling Gpihbp1 expression., (Copyright © 2019 Allan et al.)- Published
- 2019
- Full Text
- View/download PDF
47. Mutations in RELT cause autosomal recessive amelogenesis imperfecta.
- Author
-
Kim JW, Zhang H, Seymen F, Koruyucu M, Hu Y, Kang J, Kim YJ, Ikeda A, Kasimoglu Y, Bayram M, Zhang C, Kawasaki K, Bartlett JD, Saunders TL, Simmer JP, and Hu JC
- Subjects
- Consanguinity, Genotype, Germ-Line Mutation, Humans, In Situ Hybridization, Pedigree, Phenotype, RNA Splicing, Exome Sequencing, Amelogenesis Imperfecta diagnosis, Amelogenesis Imperfecta genetics, Genes, Recessive, Genetic Association Studies, Genetic Predisposition to Disease, Mutation, Receptors, Tumor Necrosis Factor genetics
- Abstract
Amelogenesis imperfecta (AI) is a collection of isolated (non-syndromic) inherited diseases affecting dental enamel formation or a clinical phenotype in syndromic conditions. We characterized three consanguineous AI families with generalized irregular hypoplastic enamel with rapid attrition that perfectly segregated with homozygous defects in a novel gene: RELT that is a member of the tumor necrosis factor receptor superfamily (TNFRSF). RNAscope in situ hybridization of wild-type mouse molars and incisors showed specific Relt mRNA expression by secretory stage ameloblasts and by odontoblasts. Relt
-/- mice generated by CRISPR/Cas9 exhibited incisor and molar enamel malformations. Relt-/- enamel had a rough surface and underwent rapid attrition. Normally unmineralized spaces in the deep enamel near the dentino-enamel junction (DEJ) were as highly mineralized as the adjacent enamel, which likely altered the mechanical properties of the DEJ. Phylogenetic analyses showed the existence of selective pressure on RELT gene outside of tooth development, indicating that the human condition may be syndromic, which possibly explains the history of small stature and severe childhood infections in two of the probands. Knowing a TNFRSF member is critical during the secretory stage of enamel formation advances our understanding of amelogenesis and improves our ability to diagnose human conditions featuring enamel malformations., (© 2018 The Authors. Clinical Genetics published by John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)- Published
- 2019
- Full Text
- View/download PDF
48. Next Generation Transgenic Rat Model Production.
- Author
-
Filipiak WE, Hughes ED, Gavrilina GB, LaForest AK, and Saunders TL
- Subjects
- Animals, CRISPR-Cas Systems, Gene Expression, Genes, Reporter, Microinjections veterinary, Models, Animal, Rats, Rats, Transgenic genetics, Genetic Engineering methods, Rats, Transgenic growth & development, Zygote growth & development
- Abstract
The next generation of new genetically engineered rat models by microinjection is described. Genome editors such as CRISPR/Cas9 have greatly increased the efficiency with which the rat genome can be modified to generate research models for biomedical research. Pronuclear microinjection of transgene DNA into rat zygotes results in random multicopy transgene integration events that use exogenous promoters to drive expression. Best practices in transgenic animal design indicate the use of precise single copy transgene integration in the genome. This ideal can be achieved by repair of CRISPR/Cas9 chromosome breaks by homology directed repair. The most effective way to achieve this type of transgenic rat model is to deliver genome modification reagents to rat zygotes by pronuclear microinjection. The keys to success in this process are to obtain fertilized eggs (zygotes) from the rat strain of choice, to purify the microinjection reagents, to deliver the reagents to the eggs by pronuclear microinjection, to use the surgical transfer of microinjected eggs to pseudopregnant rats to obtain G0 founder animals that carry the novel genetic modification. Ultimately the success of new rat models is measured by changes in gene expression as in the expression of a new reporter protein such as eGFP, Cre recombinase, or other protein of interest.
- Published
- 2019
- Full Text
- View/download PDF
49. Transgene Recombineering in Bacterial Artificial Chromosomes.
- Author
-
Zeidler MG and Saunders TL
- Subjects
- Animals, Animals, Genetically Modified, Genes, Reporter, Genome Size, Genome, Bacterial, Humans, Chromosomes, Artificial, Bacterial genetics, Genetic Engineering methods, Homologous Recombination, Transgenes
- Abstract
Bacterial Artificial Chromosome (BAC) libraries are a valuable research resource. Any one of the clones in these libraries can carry hundreds of thousands of base pairs of genetic information. Often the entire coding sequence and significant upstream and downstream regions, including regulatory elements, can be found in a single BAC clone. BACs can be put to many uses, such as to study the function of human genes in knockout mice, to drive reporter gene expression in transgenic animals, and for gene discovery. In order to use BACs for experimental purposes it is often desirable to genetically modify them by introducing reporter elements or heterologous cDNA sequences. It is not feasible to use conventional DNA cloning approaches to modify BACs due to their size and complexity, thus a specialized field "recombineering" has developed to modify BAC clones through the use of homologous recombination in bacteria with short homology regions. Genetically engineered BACs can then be used in cell culture, mouse, or rat models to study cancer, neurology, and genetics.
- Published
- 2019
- Full Text
- View/download PDF
50. Thioredoxin overexpression in both the cytosol and mitochondria accelerates age-related disease and shortens lifespan in male C57BL/6 mice.
- Author
-
Cunningham GM, Flores LC, Roman MG, Cheng C, Dube S, Allen C, Valentine JM, Hubbard GB, Bai Y, Saunders TL, and Ikeno Y
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Models, Animal, Aging physiology, Cytosol metabolism, Longevity physiology, Mitochondria metabolism, Thioredoxins metabolism
- Abstract
To investigate the role of increased levels of thioredoxin (Trx) in both the cytosol (Trx1) and mitochondria (Trx2) on aging, we have conducted a study to examine survival and age-related diseases using male mice overexpressing Trx1 and Trx2 (TXNTg × TXN2Tg). Our study demonstrated that the upregulation of Trx in both the cytosol and mitochondria in male TXNTg × TXN2Tg C57BL/6 mice resulted in a significantly shorter lifespan compared to wild-type (WT) mice. Cross-sectional pathology data showed a slightly higher incidence of neoplastic diseases in TXNTg × TXN2Tg mice than WT mice. The incidence of lymphoma, a major neoplastic disease in C57BL/6 mice, was slightly higher in TXNTg × TXN2Tg mice than in WT mice, and more importantly, the severity of lymphoma was significantly higher in TXNTg × TXN2Tg mice compared to WT mice. Furthermore, the total number of histopathological changes in the whole body (disease burden) was significantly higher in TXNTg × TXN2Tg mice compared to WT mice. Therefore, our study suggests that overexpression of Trx in both the cytosol and mitochondria resulted in deleterious effects on aging and accelerated the development of age-related diseases, especially cancer, in male C57BL/6 mice.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.