102 results on '"Lyons GE"'
Search Results
2. Expression of mef2 genes in the mouse central nervous system suggests a role in neuronal maturation
- Author
-
Lyons, GE, primary, Micales, BK, additional, Schwarz, J, additional, Martin, JF, additional, and Olson, EN, additional
- Published
- 1995
- Full Text
- View/download PDF
3. Irx4 Marks a Multipotent, Ventricular-Specific Progenitor Cell.
- Author
-
Nelson DO, Lalit PA, Biermann M, Markandeya YS, Capes DL, Addesso L, Patel G, Han T, John MC, Powers PA, Downs KM, Kamp TJ, and Lyons GE
- Subjects
- Animals, Biomarkers metabolism, Cell Line, Cell Membrane metabolism, Cell Proliferation, Cell Separation, Clone Cells, Embryonic Development, Endothelial Cells metabolism, Gene Expression Regulation, Developmental, Genes, Reporter, Mice, Mouse Embryonic Stem Cells cytology, Mouse Embryonic Stem Cells metabolism, Myocytes, Cardiac metabolism, Myocytes, Smooth Muscle cytology, Organ Specificity, Single-Cell Analysis, Time Factors, Heart Ventricles cytology, Homeodomain Proteins metabolism, Multipotent Stem Cells cytology, Multipotent Stem Cells metabolism
- Abstract
While much progress has been made in the resolution of the cellular hierarchy underlying cardiogenesis, our understanding of chamber-specific myocardium differentiation remains incomplete. To better understand ventricular myocardium differentiation, we targeted the ventricle-specific gene, Irx4, in mouse embryonic stem cells to generate a reporter cell line. Using an antibiotic-selection approach, we purified Irx4
+ cells in vitro from differentiating embryoid bodies. The isolated Irx4+ cells proved to be highly proliferative and presented Cxcr4, Pdgfr-alpha, Flk1, and Flt1 on the cell surface. Single Irx4+ ventricular progenitor cells (VPCs) exhibited cardiovascular potency, generating endothelial cells, smooth muscle cells, and ventricular myocytes in vitro. The ventricular specificity of the Irx4+ population was further demonstrated in vivo as VPCs injected into the cardiac crescent subsequently produced Mlc2v+ myocytes that exclusively contributed to the nascent ventricle at E9.5. These findings support the existence of a newly identified ventricular myocardial progenitor. This is the first report of a multipotent cardiac progenitor that contributes progeny specific to the ventricular myocardium. Stem Cells 2016;34:2875-2888., (© 2016 AlphaMed Press.)- Published
- 2016
- Full Text
- View/download PDF
4. Hepatitis C virus-cross-reactive TCR gene-modified T cells: a model for immunotherapy against diseases with genomic instability.
- Author
-
Spear TT, Riley TP, Lyons GE, Callender GG, Roszkowski JJ, Wang Y, Simms PE, Scurti GM, Foley KC, Murray DC, Hellman LM, McMahan RH, Iwashima M, Garrett-Mayer E, Rosen HR, Baker BM, and Nishimura MI
- Subjects
- Antigens, Viral immunology, CD8-Positive T-Lymphocytes immunology, Cross Reactions, Hepacivirus genetics, Hepatitis C etiology, Humans, Epitopes, T-Lymphocyte immunology, Genomic Instability, Hepacivirus immunology, Hepatitis C prevention & control, Histocompatibility Antigens Class I immunology, Immunotherapy, Receptors, Antigen, T-Cell immunology
- Abstract
A major obstacle hindering the development of effective immunity against viral infections, their associated disease, and certain cancers is their inherent genomic instability. Accumulation of mutations can alter processing and presentation of antigens recognized by antibodies and T cells that can lead to immune escape variants. Use of an agent that can intrinsically combat rapidly mutating viral or cancer-associated antigens would be quite advantageous in developing effective immunity against such disease. We propose that T cells harboring cross-reactive TCRs could serve as a therapeutic agent in these instances. With the use of hepatitis C virus, known for its genomic instability as a model for mutated antigen recognition, we demonstrate cross-reactivity against immunogenic and mutagenic nonstructural protein 3:1406-1415 and nonstructural protein 3:1073-1081 epitopes in PBL-derived, TCR-gene-modified T cells. These single TCR-engineered T cells can CD8-independently recognize naturally occurring and epidemiologically relevant mutant variants. TCR-peptide MHC modeling data allow us to rationalize how TCR structural properties accommodate recognition of certain mutated epitopes and how these substitutions impact the requirement of CD8 affinity enhancement for recognition. A better understanding of such TCRs' promiscuous behavior may allow for exploitation of these properties to develop novel, adoptive T cell-based therapies for viral infections and cancers exhibiting similar genomic instability., (© Society for Leukocyte Biology.)
- Published
- 2016
- Full Text
- View/download PDF
5. Lineage Reprogramming of Fibroblasts into Proliferative Induced Cardiac Progenitor Cells by Defined Factors.
- Author
-
Lalit PA, Salick MR, Nelson DO, Squirrell JM, Shafer CM, Patel NG, Saeed I, Schmuck EG, Markandeya YS, Wong R, Lea MR, Eliceiri KW, Hacker TA, Crone WC, Kyba M, Garry DJ, Stewart R, Thomson JA, Downs KM, Lyons GE, and Kamp TJ
- Subjects
- Animals, Cell Survival, Fibroblasts cytology, Mice, Mice, Transgenic, Myoblasts, Cardiac cytology, Transcription Factors genetics, Cell Proliferation, Cellular Reprogramming, Cellular Reprogramming Techniques methods, Fibroblasts metabolism, Myoblasts, Cardiac metabolism, Transcription Factors biosynthesis
- Abstract
Several studies have reported reprogramming of fibroblasts into induced cardiomyocytes; however, reprogramming into proliferative induced cardiac progenitor cells (iCPCs) remains to be accomplished. Here we report that a combination of 11 or 5 cardiac factors along with canonical Wnt and JAK/STAT signaling reprogrammed adult mouse cardiac, lung, and tail tip fibroblasts into iCPCs. The iCPCs were cardiac mesoderm-restricted progenitors that could be expanded extensively while maintaining multipotency to differentiate into cardiomyocytes, smooth muscle cells, and endothelial cells in vitro. Moreover, iCPCs injected into the cardiac crescent of mouse embryos differentiated into cardiomyocytes. iCPCs transplanted into the post-myocardial infarction mouse heart improved survival and differentiated into cardiomyocytes, smooth muscle cells, and endothelial cells. Lineage reprogramming of adult somatic cells into iCPCs provides a scalable cell source for drug discovery, disease modeling, and cardiac regenerative therapy., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
6. Irx4 identifies a chamber-specific cell population that contributes to ventricular myocardium development.
- Author
-
Nelson DO, Jin DX, Downs KM, Kamp TJ, and Lyons GE
- Subjects
- Animals, Mice, Mice, Inbred BALB C, Organ Specificity, Gene Expression Regulation, Developmental physiology, Heart Ventricles cytology, Heart Ventricles embryology, Homeodomain Proteins biosynthesis, Myocardium cytology, Myocardium metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Organogenesis physiology
- Abstract
Background: The ventricular myocardium is the most prominent layer of the heart, and the most important for mediating cardiac physiology. Although the ventricular myocardium is critical for heart function, the cellular hierarchy responsible for ventricle-specific myocardium development remains unresolved., Results: To determine the pattern and time course of ventricular myocardium development, we investigated IRX4 protein expression, which has not been previously reported. We identified IRX4+ cells in the cardiac crescent, and these cells were positive for markers of the first or second heart fields. From the onset of chamber formation, IRX4+ cells were restricted to the ventricular myocardium. This expression pattern persisted into adulthood. Of interest, we observed that IRX4 exhibits developmentally regulated dynamic intracellular localization. Throughout prenatal cardiogenesis, and up to postnatal day 4, IRX4 was detected in the cytoplasm of ventricular myocytes. However, between postnatal days 5–6, IRX4 translocated to the nucleus of ventricular myocytes., Conclusions: Given the ventricle-specific expression of Irx4 in later stages of heart development, we hypothesize that IRX4+ cells in the cardiac crescent represent the earliest cell population in the cellular hierarchy underlying ventricular myocardium development.
- Published
- 2014
- Full Text
- View/download PDF
7. Image-inspired 3D multiphoton excited fabrication of extracellular matrix structures by modulated raster scanning.
- Author
-
Ajeti V, Lien CH, Chen SJ, Su PJ, Squirrell JM, Molinarolo KH, Lyons GE, Eliceiri KW, Ogle BM, and Campagnola PJ
- Subjects
- Algorithms, Extracellular Matrix ultrastructure, Image Enhancement instrumentation, Image Interpretation, Computer-Assisted instrumentation, Microscopy, Confocal instrumentation, Microscopy, Fluorescence, Multiphoton instrumentation
- Abstract
Multiphoton excited photochemistry is a powerful 3D fabrication tool that produces sub-micron feature sizes. Here we exploit the freeform nature of the process to create models of the extracellular matrix (ECM) of several tissues, where the design blueprint is derived directly from high resolution optical microscopy images (e.g. fluorescence and Second Harmonic Generation). To achieve this goal, we implemented a new form of instrument control, termed modulated raster scanning, where rapid laser shuttering (10 MHz) is used to directly map the greyscale image data to the resulting protein concentration in the fabricated scaffold. Fidelity in terms of area coverage and relative concentration relative to the image data is ~95%. We compare the results to an STL approach, and find the new scheme provides significantly improved performance. We suggest the method will enable a variety of cell-matrix studies in cancer biology and also provide insight into generating scaffolds for tissue engineering.
- Published
- 2013
- Full Text
- View/download PDF
8. Spatial and temporal analysis of extracellular matrix proteins in the developing murine heart: a blueprint for regeneration.
- Author
-
Hanson KP, Jung JP, Tran QA, Hsu SP, Iida R, Ajeti V, Campagnola PJ, Eliceiri KW, Squirrell JM, Lyons GE, and Ogle BM
- Subjects
- Animals, Collagen Type I genetics, Collagen Type I metabolism, Collagen Type III metabolism, Elastin genetics, Elastin metabolism, Extracellular Matrix Proteins genetics, Fibronectins genetics, Fibronectins metabolism, Heart Ventricles embryology, Heart Ventricles metabolism, Immunohistochemistry, Mice, Pericardium embryology, Pericardium metabolism, Real-Time Polymerase Chain Reaction, Regeneration physiology, Extracellular Matrix Proteins metabolism, Heart embryology
- Abstract
The extracellular matrix (ECM) of the embryonic heart guides assembly and maturation of cardiac cell types and, thus, may serve as a useful template, or blueprint, for fabrication of scaffolds for cardiac tissue engineering. Surprisingly, characterization of the ECM with cardiac development is scattered and fails to comprehensively reflect the spatiotemporal dynamics making it difficult to apply to tissue engineering efforts. The objective of this work was to define a blueprint of the spatiotemporal organization, localization, and relative amount of the four essential ECM proteins, collagen types I and IV (COLI, COLIV), elastin (ELN), and fibronectin (FN) in the left ventricle of the murine heart at embryonic stages E12.5, E14.5, and E16.5 and 2 days postnatal (P2). Second harmonic generation (SHG) imaging identified fibrillar collagens at E14.5, with an increasing density over time. Subsequently, immunohistochemistry (IHC) was used to compare the spatial distribution, organization, and relative amounts of each ECM protein. COLIV was found throughout the developing heart, progressing in amount and organization from E12.5 to P2. The amount of COLI was greatest at E12.5 particularly within the epicardium. For all stages, FN was present in the epicardium, with highest levels at E12.5 and present in the myocardium and the endocardium at relatively constant levels at all time points. ELN remained relatively constant in appearance and amount throughout the developmental stages except for a transient increase at E16.5. Expression of ECM mRNA was determined using quantitative polymerase chain reaction and allowed for comparison of amounts of ECM molecules at each time point. Generally, COLI and COLIII mRNA expression levels were comparatively high, while COLIV, laminin, and FN were expressed at intermediate levels throughout the time period studied. Interestingly, levels of ELN mRNA were relatively low at early time points (E12.5), but increased significantly by P2. Thus, we identified changes in the spatial and temporal localization of the primary ECM of the developing ventricle. This characterization can serve as a blueprint for fabrication techniques, which we illustrate by using multiphoton excitation photochemistry to create a synthetic scaffold based on COLIV organization at P2. Similarly, fabricated scaffolds generated using ECM components, could be utilized for ventricular repair.
- Published
- 2013
- Full Text
- View/download PDF
9. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method.
- Author
-
Zhang J, Klos M, Wilson GF, Herman AM, Lian X, Raval KK, Barron MR, Hou L, Soerens AG, Yu J, Palecek SP, Lyons GE, Thomson JA, Herron TJ, Jalife J, and Kamp TJ
- Subjects
- Activins pharmacology, Bone Morphogenetic Protein 4 pharmacology, Cell Differentiation drug effects, Cell Line, Cells, Cultured, Drug Combinations, Epithelial-Mesenchymal Transition drug effects, Epithelial-Mesenchymal Transition physiology, Fibroblast Growth Factor 2 pharmacology, Humans, Intercellular Signaling Peptides and Proteins pharmacology, Myocytes, Cardiac drug effects, Pluripotent Stem Cells drug effects, Signal Transduction drug effects, Signal Transduction physiology, Cell Culture Techniques methods, Cell Differentiation physiology, Collagen, Extracellular Matrix physiology, Laminin, Myocytes, Cardiac cytology, Pluripotent Stem Cells cytology, Proteoglycans
- Abstract
Rationale: Cardiomyocytes (CMs) differentiated from human pluripotent stem cells (PSCs) are increasingly being used for cardiovascular research, including disease modeling, and hold promise for clinical applications. Current cardiac differentiation protocols exhibit variable success across different PSC lines and are primarily based on the application of growth factors. However, extracellular matrix is also fundamentally involved in cardiac development from the earliest morphogenetic events, such as gastrulation., Objective: We sought to develop a more effective protocol for cardiac differentiation of human PSCs by using extracellular matrix in combination with growth factors known to promote cardiogenesis., Methods and Results: PSCs were cultured as monolayers on Matrigel, an extracellular matrix preparation, and subsequently overlayed with Matrigel. The matrix sandwich promoted an epithelial-to-mesenchymal transition as in gastrulation with the generation of N-cadherin-positive mesenchymal cells. Combining the matrix sandwich with sequential application of growth factors (Activin A, bone morphogenetic protein 4, and basic fibroblast growth factor) generated CMs with high purity (up to 98%) and yield (up to 11 CMs/input PSC) from multiple PSC lines. The resulting CMs progressively matured over 30 days in culture based on myofilament expression pattern and mitotic activity. Action potentials typical of embryonic nodal, atrial, and ventricular CMs were observed, and monolayers of electrically coupled CMs modeled cardiac tissue and basic arrhythmia mechanisms., Conclusions: Dynamic extracellular matrix application promoted epithelial-mesenchymal transition of human PSCs and complemented growth factor signaling to enable robust cardiac differentiation.
- Published
- 2012
- Full Text
- View/download PDF
10. A coreceptor-independent transgenic human TCR mediates anti-tumor and anti-self immunity in mice.
- Author
-
Mehrotra S, Al-Khami AA, Klarquist J, Husain S, Naga O, Eby JM, Murali AK, Lyons GE, Li M, Spivey ND, Norell H, Martins da Palma T, Onicescu G, Diaz-Montero CM, Garrett-Mayer E, Cole DJ, Le Poole IC, and Nishimura MI
- Subjects
- Animals, CD3 Complex immunology, Flow Cytometry, HLA-A2 Antigen immunology, Humans, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Reverse Transcriptase Polymerase Chain Reaction, Autoimmunity, Immunotherapy, Adoptive methods, Neoplasms, Experimental immunology, Neoplasms, Experimental therapy, Receptors, Antigen, T-Cell immunology, T-Lymphocyte Subsets immunology
- Abstract
Recent advancements in T cell immunotherapy suggest that T cells engineered with high-affinity TCR can offer better tumor regression. However, whether a high-affinity TCR alone is sufficient to control tumor growth, or the T cell subset bearing the TCR is also important remains unclear. Using the human tyrosinase epitope-reactive, CD8-independent, high-affinity TCR isolated from MHC class I-restricted CD4(+) T cells obtained from tumor-infiltrating lymphocytes (TIL) of a metastatic melanoma patient, we developed a novel TCR transgenic mouse with a C57BL/6 background. This HLA-A2-restricted TCR was positively selected on both CD4(+) and CD8(+) single-positive cells. However, when the TCR transgenic mouse was developed with a HLA-A2 background, the transgenic TCR was primarily expressed by CD3(+)CD4(-)CD8(-) double-negative T cells. TIL 1383I TCR transgenic CD4(+), CD8(+), and CD4(-)CD8(-) T cells were functional and retained the ability to control tumor growth without the need for vaccination or cytokine support in vivo. Furthermore, the HLA-A2(+)/human tyrosinase TCR double-transgenic mice developed spontaneous hair depigmentation and had visual defects that progressed with age. Our data show that the expression of the high-affinity TIL 1383I TCR alone in CD3(+) T cells is sufficient to control the growth of murine and human melanoma, and the presence or absence of CD4 and CD8 coreceptors had little effect on its functional capacity.
- Published
- 2012
- Full Text
- View/download PDF
11. Imaging cardiac extracellular matrices: a blueprint for regeneration.
- Author
-
Jung JP, Squirrell JM, Lyons GE, Eliceiri KW, and Ogle BM
- Subjects
- Animals, Humans, Extracellular Matrix chemistry, Heart physiology, Myocardium cytology, Regeneration, Tissue Engineering methods
- Abstract
Once damaged, cardiac tissue does not readily repair and is therefore a primary target of regenerative therapies. One regenerative approach is the development of scaffolds that functionally mimic the cardiac extracellular matrix (ECM) to deliver stem cells or cardiac precursor populations to the heart. Technological advances in micro/nanotechnology, stem cell biology, biomaterials and tissue decellularization have propelled this promising approach forward. Surprisingly, technological advances in optical imaging methods have not been fully utilized in the field of cardiac regeneration. Here, we describe and provide examples to demonstrate how advanced imaging techniques could revolutionize how ECM-mimicking cardiac tissues are informed and evaluated., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
12. NKG2D signaling on CD8⁺ T cells represses T-bet and rescues CD4-unhelped CD8⁺ T cell memory recall but not effector responses.
- Author
-
Zloza A, Kohlhapp FJ, Lyons GE, Schenkel JM, Moore TV, Lacek AT, O'Sullivan JA, Varanasi V, Williams JW, Jagoda MC, Bellavance EC, Marzo AL, Thomas PG, Zafirova B, Polić B, Al-Harthi L, Sperling AI, and Guevara-Patiño JA
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes cytology, Cytotoxicity, Immunologic, Disease Models, Animal, Gene Expression, HIV-1 immunology, Humans, Immunity, Cellular, Influenza, Human genetics, Interferon-gamma immunology, Interferon-gamma metabolism, Interleukin-2 immunology, Interleukin-2 metabolism, Mice, Mice, Inbred C57BL, NK Cell Lectin-Like Receptor Subfamily K metabolism, T-Box Domain Proteins metabolism, T-Lymphocytes, Helper-Inducer immunology, Vaccines, DNA immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, HIV Infections immunology, Influenza, Human immunology, NK Cell Lectin-Like Receptor Subfamily K genetics, NK Cell Lectin-Like Receptor Subfamily K immunology
- Abstract
CD4-unhelped CD8(+) T cells are functionally defective T cells primed in the absence of CD4(+) T cell help. Given the co-stimulatory role of natural-killer group 2, member D protein (NKG2D) on CD8(+) T cells, we investigated its ability to rescue these immunologically impotent cells. We demonstrate that augmented co-stimulation through NKG2D during priming paradoxically rescues memory, but not effector, CD8(+) T cell responses. NKG2D-mediated rescue is characterized by reversal of elevated transcription factor T-box expressed in T cells (T-bet) expression and recovery of interleukin-2 and interferon-γ production and cytolytic responses. Rescue is abrogated in CD8(+) T cells lacking NKG2D. Augmented co-stimulation through NKG2D confers a high rate of survival to mice lacking CD4(+) T cells in a CD4-dependent influenza model and rescues HIV-specific CD8(+) T cell responses from CD4-deficient HIV-positive donors. These findings demonstrate that augmented co-stimulation through NKG2D is effective in rescuing CD4-unhelped CD8(+) T cells from their pathophysiological fate and may provide therapeutic benefits.
- Published
- 2012
- Full Text
- View/download PDF
13. Endogenous fluorescence signatures in living pluripotent stem cells change with loss of potency.
- Author
-
Squirrell JM, Fong JJ, Ariza CA, Mael A, Meyer K, Shevde NK, Roopra A, Lyons GE, Kamp TJ, Eliceiri KW, and Ogle BM
- Subjects
- Animals, Cell Differentiation drug effects, Cell Survival drug effects, Embryoid Bodies cytology, Embryoid Bodies drug effects, Glucose pharmacology, Humans, Mice, Microscopy, Pluripotent Stem Cells drug effects, Spectrometry, Fluorescence, Tretinoin pharmacology, Molecular Imaging, Pluripotent Stem Cells cytology
- Abstract
The therapeutic potential of stem cells is limited by the non-uniformity of their phenotypic state. Thus it would be advantageous to noninvasively monitor stem cell status. Driven by this challenge, we employed multidimensional multiphoton microscopy to quantify changes in endogenous fluorescence occurring with pluripotent stem cell differentiation. We found that global and cellular-scale fluorescence lifetime of human embryonic stem cells (hESC) and murine embryonic stem cells (mESC) consistently decreased with differentiation. Less consistent were trends in endogenous fluorescence intensity with differentiation, suggesting intensity is more readily impacted by nuances of species and scale of analysis. What emerges is a practical and accessible approach to evaluate, and ultimately enrich, living stem cell populations based on changes in metabolism that could be exploited for both research and clinical applications.
- Published
- 2012
- Full Text
- View/download PDF
14. Engagement of NK receptor NKG2D, but not 2B4, results in self-reactive CD8+ T cells and autoimmune vitiligo.
- Author
-
Zloza A, Lyons GE, Chlewicki LK, Kohlhapp FJ, O'Sullivan JA, Lacek AT, Moore TV, Jagoda MC, Kumar V, and Guevara-Patiño JA
- Subjects
- Animals, Antigens, CD genetics, Autoimmune Diseases metabolism, CD48 Antigen, CD8-Positive T-Lymphocytes metabolism, Ligands, Lymphocyte Activation immunology, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mutant Proteins genetics, Mutant Proteins metabolism, Oxidoreductases genetics, Oxidoreductases metabolism, Signaling Lymphocytic Activation Molecule Family, Vitiligo metabolism, Antigens, CD metabolism, Autoimmune Diseases immunology, CD8-Positive T-Lymphocytes immunology, NK Cell Lectin-Like Receptor Subfamily K metabolism, Receptors, Immunologic metabolism, Vitiligo immunology
- Abstract
In this study, we demonstrate that engagement of two different natural killer receptors (NKRs) can lead to contrasting effects in the development of self-reactive CD8+T cells and autoimmune vitiligo. Specifically, using a mouse model, we show that CD8+T-cell targeting of a melanocyte antigen, tyrosinase-related protein-1 (TRP-1) in combination with delivery of the NKG2D ligands (Rae-1ϵ or H60), results in strong CD8+T-cell responses against TRP-1 and in the development of autoimmune vitiligo. In contrast, targeting of TRP-1 in combination with delivery of CD48, the natural ligand for the NKR 2B4, leads to reduced formation of TRP-1-reactive CD8+T-cell responses and decreased development of vitiligo. These data indicate that autoimmune vitiligo is limited by insufficient signals, despite plentiful self-reactive T cells in the peripheral immune system. To our knowledge, this is the first experimental evidence supporting the role of NKRs in modulating CD8+T-cell autoimmune vitiligo. This study supports the utilization of NKR signaling as a therapeutic avenue toward prevention of vitiligo and other autoimmune diseases.
- Published
- 2011
- Full Text
- View/download PDF
15. Multiphoton flow cytometry to assess intrinsic and extrinsic fluorescence in cellular aggregates: applications to stem cells.
- Author
-
Buschke DG, Squirrell JM, Ansari H, Smith MA, Rueden CT, Williams JC, Lyons GE, Kamp TJ, Eliceiri KW, and Ogle BM
- Subjects
- Imaging, Three-Dimensional, Stem Cells physiology, Cell Aggregation, Flow Cytometry methods, Fluorescence, Stem Cells chemistry, Stem Cells metabolism
- Abstract
Detection and tracking of stem cell state are difficult due to insufficient means for rapidly screening cell state in a noninvasive manner. This challenge is compounded when stem cells are cultured in aggregates or three-dimensional (3D) constructs because living cells in this form are difficult to analyze without disrupting cellular contacts. Multiphoton laser scanning microscopy is uniquely suited to analyze 3D structures due to the broad tunability of excitation sources, deep sectioning capacity, and minimal phototoxicity but is throughput limited. A novel multiphoton fluorescence excitation flow cytometry (MPFC) instrument could be used to accurately probe cells in the interior of multicell aggregates or tissue constructs in an enhanced-throughput manner and measure corresponding fluorescent properties. By exciting endogenous fluorophores as intrinsic biomarkers or exciting extrinsic reporter molecules, the properties of cells in aggregates can be understood while the viable cellular aggregates are maintained. Here we introduce a first generation MPFC system and show appropriate speed and accuracy of image capture and measured fluorescence intensity, including intrinsic fluorescence intensity. Thus, this novel instrument enables rapid characterization of stem cells and corresponding aggregates in a noninvasive manner and could dramatically transform how stem cells are studied in the laboratory and utilized in the clinic.
- Published
- 2011
- Full Text
- View/download PDF
16. CD8 co-receptor promotes susceptibility of CD8+ T cells to transforming growth factor-β (TGF-β)-mediated suppression.
- Author
-
Zloza A, Jagoda MC, Lyons GE, Graves MC, Kohlhapp FJ, O'Sullivan JA, Lacek AT, Nishimura MI, and Guevara-Patiño JA
- Subjects
- Animals, Antibodies, Monoclonal immunology, CD8 Antigens biosynthesis, Humans, Jurkat Cells, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) antagonists & inhibitors, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) biosynthesis, Mice, Mice, Inbred C57BL, Receptors, Antigen, T-Cell, alpha-beta metabolism, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction immunology, Smad Proteins metabolism, CD8 Antigens immunology, CD8-Positive T-Lymphocytes immunology, Immune Tolerance immunology, Transforming Growth Factor beta immunology
- Abstract
CD8+ T cell function depends on a finely orchestrated balance of activation/suppression signals. While the stimulatory role of the CD8 co-receptor and pleiotropic capabilities of TGF-β have been studied individually, the influence of CD8 co-receptor on TGF-β function in CD8+ T cells is unknown. Here, we show that while CD8 enhances T cell activation, it also enhances susceptibility to TGF-β-mediated immune suppression. Using Jurkat cells expressing a full-length, truncated or no αβCD8 molecule, we demonstrate that cells expressing full-length αβCD8 were highly susceptible, αβCD8-truncated cells were partially susceptible, and CD8-deficient cells were completely resistant to suppression by TGF-β. Additionally, we determined that inhibition of Lck rendered mouse CD8+ T cells highly resistant to TGF-β suppression. Resistance was not associated with TGF-β receptor expression but did correlate with decreased Smad3 and increased Smad7 levels. These findings highlight a previously unrecognized third role for CD8 co-receptor which appears to prepare activated CD8+ T cells for response to TGF-β. Based on the important role which TGF-β-mediated suppression plays in tumor immunology, these findings unveil necessary considerations in formulation of CD8+ T cell-related cancer immunotherapy strategies.
- Published
- 2011
- Full Text
- View/download PDF
17. Pitx2c modulates cardiac-specific transcription factors networks in differentiating cardiomyocytes from murine embryonic stem cells.
- Author
-
Lozano-Velasco E, Chinchilla A, Martínez-Fernández S, Hernández-Torres F, Navarro F, Lyons GE, Franco D, and Aránega AE
- Subjects
- Animals, Cell Cycle, Cell Differentiation, Cells, Cultured, Embryonic Stem Cells metabolism, Homeodomain Proteins metabolism, Mice, Mutation, Myocytes, Cardiac metabolism, Transcription Factors metabolism, Up-Regulation, Homeobox Protein PITX2, Embryonic Stem Cells cytology, Gene Expression Regulation, Developmental, Homeodomain Proteins genetics, Myocytes, Cardiac cytology, Transcription Factors genetics
- Abstract
Aim: The knowledge of the molecular signals that control cell differentiation into cardiomyocytes is critical to apply cell-based therapies and repair an injured heart. The transcription factor Pitx2 has essential roles in the development of different organs including the heart. Although a direct role of Pitx2 in the developing myocardium has recently been reported, the molecular pathways driven by Pitx2 as well as its cardiac target genes remain largely unexplored. The aim of this study was to unravel the molecular mechanisms driven by Pitx2 during the process of cardiomyocyte differentiation in vitro in mouse embryonic stem cell-derived cardiomyocytes., Methods and Results: Pitx2c was overexpressed in the R1-embryonic stem cell line. mRNA levels and protein distribution of several specific cardiac genes were analyzed by real-time PCR and immunohistochemistry experiments in R1-embryonic stem cell-derived beating areas at different stages of in vitro differentiation. Our results show that overexpression of Pitx2c in embryonic stem cell-derived cardiomyocytes is able to dynamically upregulate several cardiac-enriched transcription factors such as Isl1, Mef2c and Gata4. Additionally, Pitx2c induces the expression of chamber-specific cardiac genes such as Tbx5, Nppa and Cx40. These data were validated in an in vivo model of Pitx2 loss of function., Conclusion: Taken together, these results demonstrate that Pitx2 plays a major role reinforcing the transcriptional program of cardiac differentiation., (Copyright © 2011 S. Karger AG, Basel.)
- Published
- 2011
- Full Text
- View/download PDF
18. Identification of RING finger protein 4 (RNF4) as a modulator of DNA demethylation through a functional genomics screen.
- Author
-
Hu XV, Rodrigues TM, Tao H, Baker RK, Miraglia L, Orth AP, Lyons GE, Schultz PG, and Wu X
- Subjects
- Animals, Base Sequence, Cell Line, DNA chemistry, DNA genetics, DNA metabolism, DNA Mismatch Repair genetics, DNA Mismatch Repair physiology, Female, Genes, Lethal, Genes, Reporter, Genes, p16, Genomics, Humans, Mice, Mice, Knockout, Nuclear Proteins deficiency, Nuclear Proteins genetics, Pregnancy, Promoter Regions, Genetic, Recombinant Proteins genetics, Recombinant Proteins metabolism, Transcription Factors deficiency, Transcription Factors genetics, Ubiquitin-Protein Ligases, DNA Methylation genetics, DNA Methylation physiology, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
DNA methylation is an important epigenetic modification involved in transcriptional regulation, nuclear organization, development, aging, and disease. Although DNA methyltransferases have been characterized, the mechanisms for DNA demethylation remain poorly understood. Using a cell-based reporter assay, we performed a functional genomics screen to identify genes involved in DNA demethylation. Here we show that RNF4 (RING finger protein 4), a SUMO-dependent ubiquitin E3-ligase previously implicated in maintaining genome stability, plays a key role in active DNA demethylation. RNF4 reactivates methylation-silenced reporters and promotes global DNA demethylation. Rnf4 deficiency is embryonic lethal with higher levels of methylation in genomic DNA. Mechanistic studies show that RNF4 interacts with and requires the base excision repair enzymes TDG and APE1 for active demethylation. This activity appears to occur by enhancing the enzymatic activities that repair DNA G:T mismatches generated from methylcytosine deamination. Collectively, our study reveals a unique function for RNF4, which may serve as a direct link between epigenetic DNA demethylation and DNA repair in mammalian cells.
- Published
- 2010
- Full Text
- View/download PDF
19. The microwell control of embryoid body size in order to regulate cardiac differentiation of human embryonic stem cells.
- Author
-
Mohr JC, Zhang J, Azarin SM, Soerens AG, de Pablo JJ, Thomson JA, Lyons GE, Palecek SP, and Kamp TJ
- Subjects
- Cell Count, Cell Line, Embryonic Stem Cells metabolism, Flow Cytometry, Gene Expression Regulation, Developmental, Humans, Myocytes, Cardiac metabolism, Myosin Light Chains metabolism, Organogenesis, Time Factors, Cell Differentiation, Cell Size, Embryo, Mammalian cytology, Embryonic Stem Cells cytology, Myocytes, Cardiac cytology
- Abstract
The differentiation of human embryonic stem cells (hESCs) into cardiomyocytes (CMs) using embryoid bodies (EBs) is relatively inefficient and highly variable. Formation of EBs using standard enzymatic disaggregation techniques results in a wide range of sizes and geometries of EBs. Use of a 3-D cuboidal microwell system to culture hESCs in colonies of defined dimensions, 100-500 microm in lateral dimensions and 120 microm in depth, enabled formation of more uniform-sized EBs. The 300 microm microwells produced highest percentage of contracting EBs, but flow cytometry for myosin light chain 2A (MLC2a) expressing cells revealed a similar percentage (approximately 3%) of cardiomyocytes formed in EBs from 100 microm to 300 microm microwells. These data, and immunolabeling with anti-MF20 and MLC2a, suggest that the smaller EBs are less likely to form contracting EBs, but those contracting EBs are relatively enriched in cardiomyocytes compared to larger EB sizes where CMs make up a proportionately smaller fraction of the total cells. We conclude that microwell-engineered EB size regulates cardiogenesis and can be used for more efficient and reproducible formation of hESC-CMs needed for research and therapeutic applications., ((c) 2009 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
20. On the road to iPS cell cardiovascular applications.
- Author
-
Kamp TJ and Lyons GE
- Subjects
- Action Potentials, Animals, Calcium Signaling, Cell Lineage, Chimerism, Fibroblasts ultrastructure, Gene Expression Regulation, Developmental, Humans, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors genetics, Kruppel-Like Transcription Factors metabolism, Myocardial Contraction, Myocytes, Cardiac ultrastructure, Octamer Transcription Factor-3 genetics, Octamer Transcription Factor-3 metabolism, Organogenesis, Pluripotent Stem Cells ultrastructure, Proto-Oncogene Proteins c-myc metabolism, SOXB1 Transcription Factors genetics, SOXB1 Transcription Factors metabolism, Cell Transdifferentiation genetics, Fibroblasts metabolism, Myocytes, Cardiac metabolism, Pluripotent Stem Cells metabolism, Tissue Engineering trends
- Published
- 2009
- Full Text
- View/download PDF
21. Relationship between CD8-dependent antigen recognition, T cell functional avidity, and tumor cell recognition.
- Author
-
Moore TV, Lyons GE, Brasic N, Roszkowski JJ, Voelkl S, Mackensen A, Kast WM, Le Poole IC, and Nishimura MI
- Subjects
- Antigens, CD analysis, Cancer Vaccines therapeutic use, Cell Line, Tumor immunology, Clone Cells immunology, Clone Cells metabolism, Cytotoxicity, Immunologic, Epitopes, T-Lymphocyte immunology, Gene Rearrangement, T-Lymphocyte, HLA-A2 Antigen immunology, Humans, Immunophenotyping, Immunotherapy, Active, Jurkat Cells, Melanoma blood, Melanoma therapy, Receptors, Antigen, T-Cell, alpha-beta genetics, Recombinant Fusion Proteins immunology, gp100 Melanoma Antigen, Antigen Presentation, Antigens, Neoplasm immunology, Cancer Vaccines immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Membrane Glycoproteins immunology, Receptors, Antigen, T-Cell, alpha-beta immunology, T-Lymphocyte Subsets immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
Effective immunotherapy using T cell receptor (TCR) gene-modified T cells requires an understanding of the relationship between TCR affinity and functional avidity of T cells. In this study, we evaluate the relative affinity of two TCRs isolated from HLA-A2-restricted, gp100-reactive T cell clones with extremely high functional avidity. Furthermore, one of these T cell clones, was CD4- CD8- indicating that antigen recognition by this clone was CD8 independent. However, when these TCRs were expressed in CD8- Jurkat cells, the resulting Jurkat cells recognized gp100:209-217 peptide loaded T2 cells and had high functional avidity, but could not recognize HLA-A2+ melanoma cells expressing gp100. Tumor cell recognition by Jurkat cells expressing these TCRs could not be induced by exogenously loading the tumor cells with the native gp100:209-217 peptide. These results indicate that functional avidity of a T cell does not necessarily correlate with TCR affinity and CD8-independent antigen recognition by a T cell does not always mean its TCR will transfer CD8-independence to other effector cells. The implications of these findings are that T cells can modulate their functional avidity independent of the affinity of their TCRs.
- Published
- 2009
- Full Text
- View/download PDF
22. An adaptable hydrogel array format for 3-dimensional cell culture and analysis.
- Author
-
Jongpaiboonkit L, King WJ, Lyons GE, Paguirigan AL, Warrick JW, Beebe DJ, and Murphy WL
- Subjects
- Animals, Cell Adhesion, Cell Count, Cell Culture Techniques instrumentation, Cell Line, Cells, Cultured, Collagen Type I chemistry, Culture Media chemistry, Endothelial Cells cytology, Humans, Materials Testing, Mesenchymal Stem Cells cytology, Mice, Myocytes, Cardiac cytology, NIH 3T3 Cells, Polyethylene Glycols chemistry, Biocompatible Materials chemistry, Cell Culture Techniques methods, Hydrogels chemistry
- Abstract
Hydrogels have been commonly used as model systems for 3-dimensional (3-D) cell biology, as they have material properties that resemble natural extracellular matrices (ECMs), and their cell-interactive properties can be readily adapted in order to address a particular hypothesis. Natural and synthetic hydrogels have been used to gain fundamental insights into virtually all aspects of cell behavior, including cell adhesion, migration, and differentiated function. However, cell responses to complex 3-D environments are difficult to adequately explore due to the large number of variables that must be controlled simultaneously. Here we describe an adaptable, automated approach for 3-D cell culture within hydrogel arrays. Our initial results demonstrate that the hydrogel network chemistry (both natural and synthetic), cell type, cell density, cell adhesion ligand density, and degradability within each array spot can be systematically varied to screen for environments that promote cell viability in a 3-D context. In a test-bed application we then demonstrate that a hydrogel array format can be used to identify environments that promote viability of HL-1 cardiomyocytes, a cell line that has not been cultured previously in 3-D hydrogel matrices. Results demonstrate that the fibronectin-derived cell adhesion ligand RGDSP improves HL-1 viability in a dose-dependent manner, and that the effect of RGDSP is particularly pronounced in degrading hydrogel arrays. Importantly, in the presence of 70mum RGDSP, HL-1 cardiomyocyte viability does not decrease even after 7 days of culture in PEG hydrogels. Taken together, our results indicate that the adaptable, array-based format developed in this study may be useful as an enhanced throughput platform for 3-D culture of a variety of cell types.
- Published
- 2008
- Full Text
- View/download PDF
23. Dynamic expression patterns of leucine-rich repeat containing protein 10 in the heart.
- Author
-
Kim KH, Kim TG, Micales BK, Lyons GE, and Lee Y
- Subjects
- Animals, Cell Nucleus chemistry, Cytoplasm chemistry, Embryonic Structures, Gene Expression Regulation, Developmental, Leucine-Rich Repeat Proteins, Mice, Muscle Proteins genetics, Proteins analysis, Proteins genetics, Sarcoplasmic Reticulum chemistry, Muscle Proteins analysis, Myocardium chemistry, Myocytes, Cardiac chemistry
- Abstract
Leucine-rich repeat containing protein 10 (LRRC10) is a heart-specific factor whose function remains unknown. Examination of the intracellular location of the gene products is a critical step in determining the biological functions of the protein. Our expression analyses in mice indicate that LRRC10 is exclusively expressed from the precardiac region in early embryos to the adult heart. LRRC10 expression is markedly elevated upon birth, suggesting its role in the embryonic as well as adult hearts. Of interest, LRRC10 exhibits dynamic intracellular expression patterns in cardiomyocytes. Cardiomyocytes from embryos and newborns show diffuse cytoplasmic and nuclear staining of LRRC10. In contrast, striking striations are observed in adult cardiomyocytes, which are colocalized with the markers for the Z-line, sarcoplasmic reticulum (SR), and transverse (T)-tubule by double immunostaining. Further investigation by electron micrographs places LRRC10 in a diad region where the SR interacts with the T-tubule that locates along the Z-line., ((c) 2007 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
24. Transplanted embryonic stem cells following mouse myocardial infarction inhibit apoptosis and cardiac remodeling.
- Author
-
Singla DK, Lyons GE, and Kamp TJ
- Subjects
- Animals, Cardiomegaly etiology, Cardiomegaly pathology, Cardiomegaly physiopathology, Cell Line, Cell Size, Collagen metabolism, Coronary Vessels surgery, Disease Models, Animal, Female, Fibrosis, Ligation, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction complications, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Regeneration, Time Factors, Apoptosis, Cardiomegaly prevention & control, Embryonic Stem Cells transplantation, Myocardial Infarction surgery, Stem Cell Transplantation, Ventricular Remodeling
- Abstract
We have previously shown that mouse embryonic stem (ES) cells transplanted following myocardial infarction (MI) differentiate into the major cell types in the heart and improve cardiac function. However, the extent of regeneration was relatively meager compared with the observed functional improvement. Therefore, we hypothesize that mechanisms in addition to regeneration contribute to the functional improvement from ES cell therapy. In this study, we examined the effect of mouse ES cells transplanted post-MI on cardiac apoptosis, fibrosis, and hypertrophy. MI was produced by left coronary artery ligation in C57BL/6 mice. Two different mouse ES cell lines, expressing enhanced green fluorescent protein and beta-galactosidase, respectively, were tested. Post-MI intramyocardial injection of 3 x 10(4) ES cells was compared with injection of medium alone. Terminal deoxynucleotidyl nick end labeling (TUNEL), immunofluorescence, and histology were used to examine the effect of transplanted ES cells on apoptosis, fibrosis, and hypertrophy. Two weeks post-MI, ES cell-transplanted hearts exhibited a significant decrease in TUNEL-stained nuclei (mean +/- SE; MI+medium = 12 +/- 1.5%; MI+ES cells = 6.6 +/- 1%, P < 0.05). TUNEL-positive nuclei were confirmed to be apoptotic by colabeling with a caspase-3 antibody. Cardiac fibrosis was 57% less in the MI+ES cell group compared with the MI + medium group (P < 0.05) as shown with Masson's trichrome staining. Picrosirius red staining confirmed a decreased amount of collagen present in the MI+ES cell group. Cardiomyocyte hypertrophy was significantly decreased following ES cell transplantation compared with medium control animals. In conclusion, transplanted mouse ES cells in the infarcted heart inhibit apoptosis, fibrosis, and hypertrophy, thereby reducing adverse remodeling.
- Published
- 2007
- Full Text
- View/download PDF
25. Evidence for functional inter-relationships between FOXP3, leukaemia inhibitory factor, and axotrophin/MARCH-7 in transplantation tolerance.
- Author
-
Muthukumarana PA, Lyons GE, Miura Y, Thompson LH, Watson T, Green CJ, Shurey S, Hess AD, Rosengard BR, and Metcalfe SM
- Subjects
- Animals, Bone Marrow Transplantation immunology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Female, Forkhead Transcription Factors metabolism, Gene Expression immunology, Graft vs Host Disease immunology, Graft vs Host Disease metabolism, Humans, Interferon-gamma metabolism, Interleukin-2 Receptor alpha Subunit immunology, Interleukins metabolism, Leukemia Inhibitory Factor metabolism, Leukemia Inhibitory Factor pharmacology, Lymphocytes drug effects, Lymphocytes immunology, Lymphocytes metabolism, Male, Mice, Mice, Inbred BALB C, Mice, Inbred CBA, Mice, Knockout, STAT3 Transcription Factor metabolism, Spleen cytology, Spleen immunology, Spleen metabolism, Suppressor of Cytokine Signaling 3 Protein, Suppressor of Cytokine Signaling Proteins genetics, Thymus Gland immunology, Thymus Gland metabolism, Transplantation Immunology genetics, Transplantation Immunology immunology, Transplantation Tolerance genetics, Ubiquitin-Protein Ligases metabolism, Forkhead Transcription Factors genetics, Leukemia Inhibitory Factor genetics, Transplantation Tolerance immunology, Ubiquitin-Protein Ligases genetics
- Abstract
In an ex vivo mouse model, regulatory transplantation tolerance is not only linked to Foxp3, but also to release of leukaemia inhibitory factor (LIF) and to expression of axotrophin (also known as MARCH-7), a putative ubiquitin E3 ligase associated with feedback control of T cell activation and of T cell-derived LIF. Given this coordinate correlation with tolerance, we now ask if Foxp3 expression is influenced by LIF or by axotrophin. In spleen cells from allo-rejected mice we found that exogenous LIF reduced interferon gamma release in response to donor antigen by 50%, but LIF had no direct effect on levels of Foxp3 protein in allo-primed cells that were either tolerant, or aggressive, for donor antigen. However, we did find an effect of axotrophin on Foxp3: in the axotrophin null mouse, thymic Foxp3 transcripts were reduced compared to axotrophin wildtype littermates. To test whether these findings in the mouse were of potential significance in man we measured transcript levels of axotrophin and LIF in peripheral blood cell samples collected for a recently published clinical study concerning haematopoietic stem cell recipients. In controls, human peripheral blood CD4+CD25+cells contained significantly more FOXP3 and axotrophin than CD4+CD25-cells. In bone marrow autograft recipients, where peripheral blood cell samples directly represent both the grafted tissue and the immune response, both FOXP3 and axotrophin negatively correlated with graft versus host disease (GVHD). These data suggest that (i) thymic Foxp3+T cell development is influenced by axotrophin; and (ii) clinical auto-GVHD inversely correlates with axotrophin transcript expression as has been previously reported for FOXP3.
- Published
- 2006
- Full Text
- View/download PDF
26. Influence of human CD8 on antigen recognition by T-cell receptor-transduced cells.
- Author
-
Lyons GE, Moore T, Brasic N, Li M, Roszkowski JJ, and Nishimura MI
- Subjects
- CD3 Complex metabolism, CD8 Antigens chemistry, CD8 Antigens genetics, CD8 Antigens immunology, Dimerization, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Genetic Vectors genetics, Humans, Interleukin-2 metabolism, Jurkat Cells, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, Mutation genetics, Protein Binding genetics, Protein Binding immunology, Receptors, Antigen, T-Cell genetics, Retroviridae genetics, T-Lymphocytes immunology, T-Lymphocytes metabolism, Transfection, Antigens, Neoplasm metabolism, CD8 Antigens metabolism, Receptors, Antigen, T-Cell metabolism
- Abstract
The CD8 coreceptor on T cells has two functions. Namely, CD8 acts to stabilize the binding of the T-cell receptor (TCR) to the peptide-MHC complex while localizing p56(lck) (lck) to the TCR/CD3 complex to facilitate early signaling events. Although both functions may be critical for efficient activation of a CTL, little is known about how the structural versus signaling roles of CD8, together with the relative strength of the TCR, influences T-cell function. We have addressed these issues by introducing full-length and truncated versions of the CD8alpha and CD8beta chains into CD8(-) Jurkat cell clones expressing cloned TCRs with known antigen specificity and relative affinities. Using a combination of antigen recognition and tetramer-binding assays, we find that the intracellular lck-binding domain of CD8 is critical for enhanced T-cell activation regardless of the relative strength of the TCR. In contrast, the extracellular domain of CD8 seems to be critical for TCRs with lower affinity but not those with higher affinity. Based on our results, we conclude that there are different requirements for CD8 to enhance T-cell function depending on the strength of its TCR.
- Published
- 2006
- Full Text
- View/download PDF
27. Functional conservation of zinc-finger homeodomain gene zfh1/SIP1 in Drosophila heart development.
- Author
-
Liu M, Su M, Lyons GE, and Bodmer R
- Subjects
- Animals, Base Sequence, DNA Primers, DNA, Complementary, Drosophila genetics, Evolution, Molecular, DNA-Binding Proteins genetics, Drosophila embryology, Drosophila Proteins genetics, Heart embryology, Repressor Proteins genetics, Zinc Fingers
- Abstract
Comparative genetic studies of diverse animal model systems have revealed that similar developmental mechanisms operate across the Metazoa. In many cases, the genes from one organism can functionally replace homologues in other phyla, a result consistent with a high degree of evolutionarily conserved gene function. We investigated functional conservation among the Drosophila zinc-finger homeodomain protein 1 (zfh1) and its mouse functional homologue Smad-interacting protein 1 (SIP1). Northern blot analyses of SIP1 expression patterns detected three novel variants (8.3, 2.7, and 1.9 kb) in addition to the previously described 5.3 kb SIP1 transcript. The two shorter novel SIP1 transcripts were encountered only in developing embryos and both lacked zinc-finger clusters or homeodomain regions. The SIP1 transcripts showed complex embryonic expression patterns consistent with that observed for Drosophila zfh1. They were highly expressed in the developing nervous systems and in a number of mesoderm-derived tissues including lungs, heart, developing myotomes, skeletal muscle, and visceral smooth muscle. The expression of the mammalian 5.3 kb SIP1 transcript in Drosophila zfh1 null mutant embryos completely restored normal heart development in the fly, demonstrating their functional equivalence in cardiogenic pathways. Our present data, together with the previously described heart defects associated with both SIP1 and Drosophila zfh1 mutations, solidify the conclusion that the zfh1 family members participate in an evolutionary conserved program of metazoan cardiogenesis.
- Published
- 2006
- Full Text
- View/download PDF
28. Pitx2c overexpression promotes cell proliferation and arrests differentiation in myoblasts.
- Author
-
Martínez-Fernandez S, Hernández-Torres F, Franco D, Lyons GE, Navarro F, and Aránega AE
- Subjects
- Animals, Cell Cycle genetics, Cell Proliferation, Cyclin D1 genetics, Cyclin D2, Cyclins genetics, Genes, myc genetics, Homeodomain Proteins analysis, Homeodomain Proteins genetics, Mice, Muscle Fibers, Skeletal chemistry, Muscle Fibers, Skeletal metabolism, Myoblasts, Skeletal chemistry, Myoblasts, Skeletal metabolism, Protein Isoforms analysis, Protein Isoforms genetics, Protein Isoforms physiology, Transcription Factors analysis, Transcription Factors genetics, Transcription, Genetic, Homeobox Protein PITX2, Cell Differentiation, Gene Expression Regulation, Developmental, Homeodomain Proteins physiology, Muscle Development genetics, Myoblasts, Skeletal cytology, Transcription Factors physiology
- Abstract
Pitx2 is a paired-related homeobox gene that has been shown to play a central role during development. In the mouse, there are three isoforms, Pitx2a, b, and c, which differ only in their amino terminal regions. Pitx2 is expressed in myotomes, myoblasts, and myofibers and may be involved in muscle patterning. However, the mechanism by which Pitx2 acts in muscle cell lineages as well as the distinct functions of the individual isoforms have not been investigated. In this study, we used Sol8 myoblasts to investigate the function of Pitx2 in skeletal myogenesis. We found that Pitx2c is the main Pitx2 isoform present in Sol8 myoblasts. Overexpression of Pitx2c in Sol8 myoblasts inhibited myocyte differentiation and myotube formation. Furthermore, Sol8 cells overexpressing Pitx2c maintained high proliferative capacity and a significant up-regulation of the cell cycle genes cyclin D1, cyclin D2, and c-myc. Gene expression analysis for Pax3 and the s MyoD and myogenin showed that Pitx2c-overexpression caused Sol8 cells to remain as myoblasts, in an undifferentiated myogenic state. Furthermore, down-regulation of the muscle-specific genes sTnI and MyHC3 demonstrated that Sol8-overexpressing Pitx2c myoblasts failed to reach terminal differentiation. This study sheds light on previously unknown functions of the Pitx2c isoform in balancing proliferation vs. differentiation in a myogenic cell line., ((c) 2006 Wiley-Liss, Inc.)
- Published
- 2006
- Full Text
- View/download PDF
29. T-cell receptor tetramer binding or the lack there of does not necessitate antigen reactivity in T-cell receptor transduced T cells.
- Author
-
Lyons GE, Roszkowski JJ, Man S, Yee C, Kast WM, and Nishimura MI
- Subjects
- Cell Line, Tumor, Epitopes, Genetic Vectors, HLA-A2 Antigen, Humans, Jurkat Cells, Receptors, Antigen, T-Cell classification, Retroviridae genetics, Transduction, Genetic, HLA-A Antigens metabolism, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism
- Abstract
Genetic transfer of T-cell receptor (TCR) chains provides a means of transferring tumor antigen specificity onto an alternate T-cell population. To determine which tumor reactive TCRs are best suitable for such adoptive transfer, careful evaluation of the resulting TCR modified populations need to be performed. We have previously cloned, and expressed TCRs from melanoma, EBV, HCV, and HPV reactive T-cell clones and found that several routine indicators of T-cell function do not always predict the relative strength of a TCR. Using a combination of tetramer binding assays and antigen recognition assays, we identified TCRs that fall into three classes. One class of TCR did not bind tetramers yet resulted in cells with high avidity for antigen. A second TCR class bound tetramer but did not secrete cytokines in response to antigen. Finally, the third class of TCRs bound tetramer and reacted to antigen as would be expected. We conclude that tetramer binding is not always a good indicator of the function of a cloned TCR or the avidity of a TCR gene modified T cell. Furthermore, our data indicate that the use of tetramer binding alone to identify antigen reactive TCRs may result in the exclusion of TCRs that may be highly reactive or cross reactive to the relevant tumor antigen.
- Published
- 2006
- Full Text
- View/download PDF
30. Identification of a hepatitis C virus-reactive T cell receptor that does not require CD8 for target cell recognition.
- Author
-
Callender GG, Rosen HR, Roszkowski JJ, Lyons GE, Li M, Moore T, Brasic N, McKee MD, and Nishimura MI
- Subjects
- Cells, Cultured, Humans, CD8 Antigens physiology, Hepacivirus immunology, Receptors, Antigen, T-Cell physiology
- Abstract
Hepatitis C virus (HCV) has been reported to elicit B and T cell immunity in infected patients. Despite the presence of antiviral immunity, many patients develop chronic infections leading to cirrhosis, hepatocellular carcinoma, and liver failure that can require transplantation. We have previously described the presence of HLA-A2-restricted, HCV NS3-reactive cytotoxic T lymphocytes (CTL) in the blood of HLA-A2- liver transplantation patients that received an HLA-A2+ liver allograft. These T cells are analogous to the "allospecific" T cells that have been described in hematopoietic stem cell transplantation patients. It has been speculated that allospecific T cells express high-affinity T cell receptors (TCRs). To determine if our HCV-reactive T cells expressed TCRs with relatively high affinity for antigen, we identified and cloned a TCR from an allospecific HLA-A2-restricted, HCV:NS3:1406-1415-reactive CD8+ T cell clone and expressed this HCV TCR in Jurkat cells. Tetramer binding to HCV TCR-transduced Jurkat cells required CD8 expression, whereas antigen recognition did not. In conclusion, based on the reactivity of the TCR-transduced Jurkat cells, we have identified a TCR that transfers anti-HCV reactivity to alternate effectors. These data suggest this high affinity HCV-specific TCR might have potential new immunotherapic implications.
- Published
- 2006
- Full Text
- View/download PDF
31. Transplantation of embryonic stem cells into the infarcted mouse heart: formation of multiple cell types.
- Author
-
Singla DK, Hacker TA, Ma L, Douglas PS, Sullivan R, Lyons GE, and Kamp TJ
- Subjects
- Animals, Cell Differentiation, Electrocardiography, Endothelium, Vascular cytology, Female, Male, Mice, Mice, Inbred C57BL, Muscle, Smooth, Vascular cytology, Myocytes, Cardiac physiology, beta-Galactosidase genetics, beta-Galactosidase metabolism, Embryo, Mammalian cytology, Myocardial Infarction pathology, Myocardium cytology, Stem Cell Transplantation methods
- Abstract
Initial studies have suggested that transplantation of embryonic stem (ES) cells following myocardial infarction (MI) in animal models is beneficial; however, the mechanism of benefit is largely unknown. The present study investigated the fate of mouse ES cells transplanted post-MI to determine if the ES cells give rise to the range of major cell types present in the native myocardium. MI was produced by coronary artery ligation in C57BL/6 mice. Two different mouse ES cell lines, expressing eGFP and beta-galactosidase, respectively, were tested. Post-MI intramyocardial injection of 3 x 10(4) ES cells was compared to injection of media alone. Histochemistry and immunofluorescence were used to track the transplanted ES cells and identify the resulting cell types. Echocardiography assessed the cardiac size and function in a blinded fashion. Two weeks post-MI, engraftment of the transplanted ES cells was demonstrated by eGFP or beta-galactosidase-positive cells in the infarct region without evidence for tumor formation. Co-immunolabeling demonstrated that the transplanted ES cells had become cardiomyocytes, vascular smooth muscle, and endothelial cells. Echocardiographic analysis showed that ES cell transplantation resulted in reduced post-MI remodeling of the heart and improved cardiac function. In conclusion, transplanted mouse ES cells can regenerate infarcted myocardium in part by becoming cardiomyocytes, vascular smooth muscle, and endothelial cells that result in an improvement in cardiac structure and function. Therefore, ES cells hold promise for myocardial cellular therapy.
- Published
- 2006
- Full Text
- View/download PDF
32. CAR-1, a protein that localizes with the mRNA decapping component DCAP-1, is required for cytokinesis and ER organization in Caenorhabditis elegans embryos.
- Author
-
Squirrell JM, Eggers ZT, Luedke N, Saari B, Grimson A, Lyons GE, Anderson P, and White JG
- Subjects
- Animals, Caenorhabditis elegans embryology, Caenorhabditis elegans genetics, Caenorhabditis elegans Proteins genetics, Cytokinesis, Cytoplasm metabolism, Embryo, Nonmammalian embryology, Endoribonucleases genetics, Gene Expression Regulation, Protein Transport, RNA Caps genetics, RNA Caps metabolism, RNA Interference, RNA-Binding Proteins genetics, Time Factors, Caenorhabditis elegans cytology, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Embryo, Nonmammalian cytology, Embryo, Nonmammalian metabolism, Endoplasmic Reticulum metabolism, Endoribonucleases metabolism, RNA-Binding Proteins metabolism
- Abstract
The division of one cell into two requires the coordination of multiple components. We describe a gene, car-1, whose product may provide a link between disparate cellular processes. Inhibition of car-1 expression in Caenorhabditis elegans embryos causes late cytokinesis failures: cleavage furrows ingress but subsequently regress and the spindle midzone fails to form, even though midzone components are present. The localized accumulation of membrane that normally develops at the apex of the cleavage furrow during the final phase of cytokinesis does not occur and organization of the endoplasmic reticulum is aberrant, indicative of a disruption in membrane trafficking. The car-1 gene has homologues in a number of species, including proteins that associate with RNA binding proteins. CAR-1 localizes to P-granules (germ-line specific ribonucleoprotein particles) and discrete, developmentally regulated cytoplasmic foci. These foci also contain DCAP-1, a protein involved in decapping mRNAs. Thus, CAR-1, a protein likely to be associated with RNA metabolism, plays an essential role in the late stage of cytokinesis, suggesting a novel link between RNA, membrane trafficking and cytokinesis in the C. elegans embryo.
- Published
- 2006
- Full Text
- View/download PDF
33. Jumonji regulates cardiomyocyte proliferation via interaction with retinoblastoma protein.
- Author
-
Jung J, Kim TG, Lyons GE, Kim HR, and Lee Y
- Subjects
- Animals, Cell Cycle physiology, Cells, Cultured, Cyclin D1 metabolism, Cyclin D2, Cyclins metabolism, Embryo, Mammalian anatomy & histology, Embryo, Mammalian physiology, Gene Expression Regulation, Genes, Reporter, Humans, Mice, Myocytes, Cardiac cytology, Nerve Tissue Proteins genetics, Phenotype, Polycomb Repressive Complex 2, Promoter Regions, Genetic, Protein Structure, Tertiary, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Retinoblastoma Protein genetics, Cell Proliferation, Myocytes, Cardiac physiology, Nerve Tissue Proteins metabolism, Retinoblastoma Protein metabolism
- Abstract
Jumonji (JMJ) can function as a transcriptional repressor and plays critical roles in embryonic development including heart development in mice. Although JMJ has been suggested to play a role in cell growth, the molecular mechanisms have not been resolved. The present data demonstrate that JMJ interacts with the retinoblastoma protein (Rb), one of the master regulatory genes of cell cycle. JMJ potentiates the repression function of Rb on E2F activities, leading to reduced cell cycle progression. The transcriptional repression domain of JMJ is critical for the interaction with Rb as well as repression of cell cycle. The physiological relevance of the association between Rb and JMJ was assessed in cardiomyocytes. Primary cardiomyocytes cultured from homozygous jmj knock-out mouse embryos (jmj mutants) show increased cell mitosis in a cardiomyocyte-specific manner. Reporter gene analyses demonstrate that promoter activities of cyclin D1, cyclin D2, and Cdc2 are up-regulated in jmj mutant cardiomyocytes. These data suggest that JMJ down-regulates the cell growth via interaction with Rb, which would provide important insights into the cardiac defects observed in jmj mutant mice.
- Published
- 2005
- Full Text
- View/download PDF
34. Simultaneous generation of CD8+ and CD4+ melanoma-reactive T cells by retroviral-mediated transfer of a single T-cell receptor.
- Author
-
Roszkowski JJ, Lyons GE, Kast WM, Yee C, Van Besien K, and Nishimura MI
- Subjects
- Cell Line, Tumor, Epitopes, T-Lymphocyte immunology, Gene Transfer Techniques, Genetic Vectors genetics, HLA-A2 Antigen immunology, Humans, Jurkat Cells, Lymphocytes, Tumor-Infiltrating immunology, Melanoma genetics, Melanoma therapy, Monophenol Monooxygenase immunology, Receptors, Antigen, T-Cell genetics, Retroviridae genetics, Transduction, Genetic, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive methods, Melanoma immunology, Receptors, Antigen, T-Cell immunology
- Abstract
Adoptive immunotherapy of cancer requires the generation of large numbers of tumor antigen-reactive T cells for transfer into cancer patients. Genes encoding tumor antigen-specific T-cell receptors can be introduced into primary human T cells by retroviral mediated gene transfer as a potential method of providing any patient with a source of autologous tumor-reactive T cells. A T-cell receptor-specific for a class I MHC (HLA-A2)-restricted epitope of the melanoma antigen tyrosinase was isolated from a CD4(+) tumor-infiltrating lymphocyte (TIL 1383I) and introduced into normal human peripheral blood lymphocytes by retroviral transduction. T-cell receptor-transduced T cells secreted various cytokines when cocultured with tyrosinase peptide-loaded antigen-presenting cells as well as melanoma cells in an HLA-A2-restricted manner, and could also lyse target cells. Furthermore, T-cell clones isolated from these cultures showed both CD8(+) and CD4(+) transduced T cells could recognize HLA-A2(+) melanoma cells, giving us the possibility of engineering class I MHC-restricted effector and T helper cells against melanoma. The ability to confer class I MHC-restricted tumor cell recognition to CD4(+) T cells makes the TIL 1383I TCR an attractive candidate for T-cell receptor gene transfer-based immunotherapy.
- Published
- 2005
- Full Text
- View/download PDF
35. Leukaemia inhibitory factor (LIF) is functionally linked to axotrophin and both LIF and axotrophin are linked to regulatory immune tolerance.
- Author
-
Metcalfe SM, Muthukumarana PA, Thompson HL, Haendel MA, and Lyons GE
- Subjects
- Animals, Cell Division physiology, Cell Separation, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Leukemia Inhibitory Factor, Mice, Mice, Inbred BALB C, Mice, Transgenic, Spleen cytology, Thymus Gland cytology, Immune Tolerance physiology, Interleukin-6 physiology
- Abstract
Axotrophin (axot) is a newly characterised stem cell gene and mice that lack axotrophin are viable and fertile, but show premature neural degeneration and defective development of the corpus callosum. By comparing axot+/+, axot+/- and axot-/- littermates, we now show that axotrophin is also involved in immune regulation. Both T cell proliferation and T cell-derived leukaemia inhibitory factor (LIF) were suppressed by axotrophin in a gene-dose-dependent manner. Moreover, a role for axotrophin in the feedback regulation of LIF is implicated. This is the first evidence that fate determination mediated by LIF maybe qualified by axotrophin.
- Published
- 2005
- Full Text
- View/download PDF
36. The Polycomb Ezh2 methyltransferase regulates muscle gene expression and skeletal muscle differentiation.
- Author
-
Caretti G, Di Padova M, Micales B, Lyons GE, and Sartorelli V
- Subjects
- Animals, Cell Differentiation physiology, Chromatin metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Enhancer of Zeste Homolog 2 Protein, Erythroid-Specific DNA-Binding Factors, Extremities embryology, Histone Deacetylase 1, Histone Deacetylases genetics, Histone Deacetylases metabolism, Histone-Lysine N-Methyltransferase, Lysine metabolism, Methylation, Methyltransferases genetics, Methyltransferases metabolism, Mice, Mice, Inbred Strains, MyoD Protein genetics, MyoD Protein metabolism, Polycomb Repressive Complex 2, Protein Structure, Tertiary, Proteins genetics, RNA, Small Interfering, Regulatory Sequences, Nucleic Acid, Serum Response Factor genetics, Serum Response Factor metabolism, Transcription Factors genetics, Transcription Factors metabolism, Transcriptional Activation, YY1 Transcription Factor, Gene Expression Regulation, Developmental, Muscle, Skeletal cytology, Muscle, Skeletal physiology, Proteins metabolism
- Abstract
The Ezh2 protein endows the Polycomb PRC2 and PRC3 complexes with histone lysine methyltransferase (HKMT) activity that is associated with transcriptional repression. We report that Ezh2 expression was developmentally regulated in the myotome compartment of mouse somites and that its down-regulation coincided with activation of muscle gene expression and differentiation of satellite-cell-derived myoblasts. Increased Ezh2 expression inhibited muscle differentiation, and this property was conferred by its SET domain, required for the HKMT activity. In undifferentiated myoblasts, endogenous Ezh2 was associated with the transcriptional regulator YY1. Both Ezh2 and YY1 were detected, with the deacetylase HDAC1, at genomic regions of silent muscle-specific genes. Their presence correlated with methylation of K27 of histone H3. YY1 was required for Ezh2 binding because RNA interference of YY1 abrogated chromatin recruitment of Ezh2 and prevented H3-K27 methylation. Upon gene activation, Ezh2, HDAC1, and YY1 dissociated from muscle loci, H3-K27 became hypomethylated and MyoD and SRF were recruited to the chromatin. These findings suggest the existence of a two-step activation mechanism whereby removal of H3-K27 methylation, conferred by an active Ezh2-containing protein complex, followed by recruitment of positive transcriptional regulators at discrete genomic loci are required to promote muscle gene expression and cell differentiation.
- Published
- 2004
- Full Text
- View/download PDF
37. Analysis of histology specimens using lifetime multiphoton microscopy.
- Author
-
Eliceiri KW, Fan CH, Lyons GE, and White JG
- Subjects
- Animals, Caenorhabditis elegans metabolism, Carbocyanines, Cell Nucleus metabolism, Cytoplasm metabolism, Cytoskeletal Proteins metabolism, DNA metabolism, Feasibility Studies, Indoles, Caenorhabditis elegans cytology, Caenorhabditis elegans embryology, Cell Nucleus ultrastructure, Cytoplasm ultrastructure, Microscopy, Confocal methods, Microscopy, Fluorescence, Multiphoton methods
- Abstract
Observations of cells or tissues with fluorescence microscopy can provide unique insights into cellular physiology and structure. Such information may reveal the pathological state of a tissue to the physician or information on cytoskeletal dynamics to the research scientist. However, problems of overlapping spectra, low signal, and light scatter impose serious limitations on what can be achieved in practice with fluorescence microscopy. These problems can be addressed in part by the development of new imaging modalities that make maximum use of the information present in the fluorescence signal. We describe the application of a new technology to the study of standard histological pathology specimens: a multiphoton excitation fluorescence microscope that incorporates a novel, photon-counting detector that measures the excited-state lifetimes of fluorescent probes. In initial investigations, we have applied this system to the observation of C. elegans embryos and primate histology specimens, with the objective of identifying potentially diagnostic signatures. Our findings demonstrate that lifetime multiphoton microscopy has considerable potential as a diagnostic tool for pathological investigations., ((c) 2003 Society of Photo-Optical Instrumentation Engineers.)
- Published
- 2003
- Full Text
- View/download PDF
38. Essential role for NFI-C/CTF transcription-replication factor in tooth root development.
- Author
-
Steele-Perkins G, Butz KG, Lyons GE, Zeichner-David M, Kim HJ, Cho MI, and Gronostajski RM
- Subjects
- Alveolar Process abnormalities, Alveolar Process growth & development, Animals, CCAAT-Enhancer-Binding Proteins deficiency, CCAAT-Enhancer-Binding Proteins genetics, Female, Gene Expression Regulation, Developmental, Gene Targeting, Incisor abnormalities, Incisor growth & development, Male, Mice, Mice, Knockout, Molar abnormalities, Molar growth & development, NFI Transcription Factors, Odontogenesis genetics, Odontogenesis physiology, Pregnancy, Tooth Root abnormalities, Transcription Factors deficiency, Transcription Factors genetics, CCAAT-Enhancer-Binding Proteins physiology, Tooth Root growth & development, Transcription Factors physiology
- Abstract
The mammalian tooth forms by a series of reciprocal epithelial-mesenchymal interactions. Although several signaling pathways and transcription factors have been implicated in regulating molar crown development, relatively little is known about the regulation of root development. Four genes encoding nuclear factor I (NFI) transcription-replication proteins are present in the mouse genome: Nfia, Nfib, Nfic, and NFIX: In order to elucidate its physiological role(s), we disrupted the Nfic gene in mice. Heterozygous animals appear normal, whereas Nfic(-/-) mice have unique tooth pathologies: molars lacking roots, thin and brittle mandibular incisors, and weakened abnormal maxillary incisors. Feeding in Nfic(-/-) mice is impaired, resulting in severe runting and premature death of mice reared on standard laboratory chow. However, a soft-dough diet mitigates the feeding impairment and maintains viability. Although Nfic is expressed in many organ systems, including the developing tooth, the tooth root development defects were the prominent phenotype. Indeed, molar crown development is normal, and well-nourished Nfic(-/-) animals are fertile and can live as long as their wild-type littermates. The Nfic mutation is the first mutation described that affects primarily tooth root formation and should greatly aid our understanding of postnatal tooth development.
- Published
- 2003
- Full Text
- View/download PDF
39. DSCAM: an endogenous promoter drives expression in the developing CNS and neural crest.
- Author
-
Barlow GM, Lyons GE, Richardson JA, Sarnat HB, and Korenberg JR
- Subjects
- 3T3 Cells, Animals, COS Cells, Cell Adhesion Molecules, Cell Line, Genes, Reporter, Humans, In Situ Hybridization, Lac Operon, Membrane Proteins, Mice, Mice, Transgenic, Models, Genetic, Neurons metabolism, Quail, Transfection, Transgenes, Central Nervous System embryology, Neural Crest embryology, Promoter Regions, Genetic, Protein Biosynthesis, Proteins genetics
- Abstract
The development of central nervous system (CNS) neuronal networks involves processes including neuroblast migration, axonal pathfinding, and synaptogenesis. To evaluate the role of the axonal guidance molecule DSCAM in CNS connectivity, we generated a lacZ reporter construct, Pr1.8-betagal, containing a 1.8kb fragment of the human DSCAM promoter region, and analyzed its expression in four E12.5 transgenic mouse embryos. We found that Pr1.8-betagal drives lacZ expression in the choroid plexus and roof of the fourth ventricle, the floor plate of the fourth ventricle, pons and medulla oblongata, and the eye, limb buds, and dorsal root ganglion. This recapitulates a subset of DSCAM expression as demonstrated by in situ hybridization, supporting this 1.8kb fragment as a component of the endogenous DSCAM promoter. The Pr1.8-betagal expression pattern supports a role for DSCAM in CNS development, providing an endogenous promoter to investigate the contribution of DSCAM to Down syndrome neural defects.
- Published
- 2002
- Full Text
- View/download PDF
40. Isolation and characterization of the mouse ortholog of the Fukuyama-type congenital muscular dystrophy gene.
- Author
-
Horie M, Kobayashi K, Takeda S, Nakamura Y, Lyons GE, and Toda T
- Subjects
- Alternative Splicing, Amino Acid Sequence, Animals, Base Sequence, Chromosome Mapping, DNA, Complementary analysis, Embryonic and Fetal Development, Gene Expression, In Situ Hybridization, Mice, Molecular Sequence Data, Muscular Dystrophies congenital, Proteins isolation & purification, Tissue Distribution, Transferases, Muscular Dystrophies genetics, Proteins genetics
- Abstract
Fukuyama-type congenital muscular dystrophy (FCMD) is a severe autosomal-recessive muscular dystrophy accompanied by brain malformation. Previously, we identified the gene responsible for FCMD through positional cloning. Here we report the isolation of its murine ortholog, Fcmd. The predicted amino acid sequence of murine fukutin protein encoded by Fcmd is 90% identical to that of its human counterpart. Radiation hybrid mapping localized the gene to 2.02 cR telomeric to D4Mit272 on chromosome 4. Northern blot analysis revealed ubiquitous expression of Fcmd in adult mouse tissues. Through in situ hybridization, we observed a wide distribution of Fcmd expression throughout embryonic development, most predominantly in the central and peripheral nervous systems. We also detected high Fcmd expression in the ventricular zone of proliferating neurons at 13.5 days post-coitum. Brain malformation in FCMD patients is thought to result from defective neuronal migration. Our data suggest that neuronally expressed Fcmd is likely to be important in the development of normal brain structure.
- Published
- 2002
41. Mammalian DSCAMs: roles in the development of the spinal cord, cortex, and cerebellum?
- Author
-
Barlow GM, Micales B, Chen XN, Lyons GE, and Korenberg JR
- Subjects
- Alternative Splicing, Animals, Brain metabolism, Cell Adhesion Molecules genetics, Cell Adhesion Molecules physiology, Cerebellum embryology, Cerebellum growth & development, Cerebellum metabolism, Cerebral Cortex embryology, Cerebral Cortex growth & development, Cerebral Cortex metabolism, DNA, Complementary analysis, Down Syndrome etiology, Humans, In Situ Hybridization, Kinetics, Membrane Proteins, Mice, Proteins genetics, Proteins physiology, RNA, Messenger biosynthesis, Spinal Cord growth & development, Spinal Cord metabolism, Brain embryology, Brain growth & development, Cell Adhesion Molecules metabolism, Proteins metabolism, Spinal Cord embryology
- Abstract
Central nervous system (CNS) development involves neural patterning, neuronal and axonal migrations, and synapse formation. DSCAM, a chromosome 21 axon guidance molecule, is expressed by CNS neurons during development and throughout adult life. We now report that DSCAM and its chromosome 11 paralog DSCAML1 exhibit inverse ventral-dorsal expression patterns in the developing spinal cord and distinct, partly inverse, expression patterns in the developing cortex, beginning in the Cajal-Retzius cells. In the adult cortex, DSCAM predominates in layer 3/5 pyramidal cells and DSCAML1 predominates in layer 2 granule cells. In the cerebellum, DSCAM is stronger in the Purkinje cells and DSCAML1 in the granule cells. Finally, we find that the predicted DSCAML1 protein contains 60 additional N-terminal amino acids which may contribute to its distinct expression pattern and putative function. We propose that the DSCAMs comprise novel elements of the pathways mediating dorsal-ventral patterning and cell-fate specification in the developing CNS., ((c) 2002 Elsevier Science (USA).)
- Published
- 2002
- Full Text
- View/download PDF
42. Characterization of murine BATF: a negative regulator of activator protein-1 activity in the thymus.
- Author
-
Williams KL, Nanda I, Lyons GE, Kuo CT, Schmid M, Leiden JM, Kaplan MH, and Taparowsky EJ
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Basic-Leucine Zipper Transcription Factors, Cell Differentiation, Cells, Cultured, Cloning, Molecular, Gene Expression Regulation, Developmental, Genes, Reporter genetics, In Situ Hybridization, In Situ Hybridization, Fluorescence, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Sequence Data, Organ Specificity, Physical Chromosome Mapping, RNA, Messenger genetics, RNA, Messenger metabolism, Spleen cytology, Spleen immunology, T-Lymphocytes cytology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Thymus Gland cytology, Thymus Gland immunology, Transcription Factor AP-1 chemistry, Transcription Factor AP-1 genetics, Transcription Factors chemistry, Transcription Factors genetics, Transcriptional Activation, Thymus Gland metabolism, Transcription Factor AP-1 antagonists & inhibitors, Transcription Factor AP-1 metabolism, Transcription Factors metabolism
- Abstract
BATF belongs to the AP-1/ATF superfamily of transcription factors and forms heterodimers with Jun proteins to bind AP-1 consensus DNA. Unlike Fos/Jun heterodimers which stimulate gene transcription, BATF/Jun heterodimers are transcriptionally inert and inhibit biological processes that are associated with the overstimulation of AP-1 activity. Here, we describe the murine BATF cDNA and genomic clones and map the BATF locus to chromosome 12 D2-3. Using in situ hybridization of BATF mRNA, we show that BATF gene expression is highly restricted, with the most prominent signals detected in the thymus. BATF mRNA levels are regulated differentially during discrete stages of T cell development and are up-regulated following activation of T cells in the periphery. To demonstrate the impact of BATF on AP-1 activity in vivo, AP-1 luciferase reporter mice were crossed to transgenic mice overexpressing BATF exclusively in thymic T cells. Results show that elevated levels of BATF protein correlate with reduced transactivation by AP-1. Since the differential regulation of AP-1 activity is linked to key transitions in the developing immune system, our observations support a critical role for BATF in determining the overall level of AP-1 activity, and thus AP-1 target gene expression, in specific T cell subtypes.
- Published
- 2001
- Full Text
- View/download PDF
43. In situ hybridization: use of 35S-labeled probes on paraffin tissue sections.
- Author
-
Micales BK and Lyons GE
- Subjects
- Acetic Anhydrides pharmacology, Animals, Diethyl Pyrocarbonate pharmacology, Embryo, Mammalian metabolism, Embryo, Mammalian pathology, Embryo, Nonmammalian, Endopeptidase K metabolism, Ethanolamines pharmacology, Nucleic Acid Hybridization, Paraffin chemistry, RNA, Messenger metabolism, Ribonucleases metabolism, Time Factors, In Situ Hybridization methods, Molecular Probes, Sulfur Radioisotopes pharmacology
- Abstract
The following protocol is for radioactive in situ hybridization detection of RNA using paraffin-embedded tissue sections on glass microscope slides. Steps taken to inhibit RNase activity such as diethyl pyrocarbonate (DEPC) treatment of solutions and baked glassware are unnecessary. The tissue is fixed using 4% paraformaldehyde, hybridized with (35)S-labeled RNA probes, and exposed to nuclear-track emulsion. The entire procedure takes 2-3 days prior to autoradiography. The time required for autoradiography is variable with an average time of 10 days. Parameters that affect the length of the autoradiography include: (1) number of copies of mRNA in the tissue, (2) incorporation of label into the probe, and (3) amount of background signal. Additional steps involved in the autoradiography process, including development of the emulsion, cleaning of the microscope slides, counterstaining of the tissue, and mounting coverslips on the microscope slides, are discussed. In addition, a general guide to the interpretation of the in situ results is provided., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
44. Down syndrome congenital heart disease: a narrowed region and a candidate gene.
- Author
-
Barlow GM, Chen XN, Shi ZY, Lyons GE, Kurnit DM, Celle L, Spinner NB, Zackai E, Pettenati MJ, Van Riper AJ, Vekemans MJ, Mjaatvedt CH, and Korenberg JR
- Subjects
- Blotting, Southern, Cell Adhesion, Cell Adhesion Molecules genetics, Cell Adhesion Molecules physiology, Child, Preschool, Chromosomes, Artificial, Bacterial, Chromosomes, Human, Pair 21, Facies, Female, Genotype, Humans, In Situ Hybridization, Fluorescence, Infant, Karyotyping, Male, Membrane Proteins, Models, Genetic, Phenotype, Pregnancy, Proteins chemistry, Proteins metabolism, Chromosome Mapping, Down Syndrome complications, Down Syndrome genetics, Heart Defects, Congenital complications, Heart Defects, Congenital genetics, Proteins genetics
- Abstract
Purpose: Down syndrome (DS) is a major cause of congenital heart disease (CHD) and the most frequent known cause of atrioventricular septal defects (AVSDs). Molecular studies of rare individuals with CHD and partial duplications of chromosome 21 established a candidate region that included D21S55 through the telomere. We now report human molecular and cardiac data that narrow the DS-CHD region, excluding two candidate regions, and propose DSCAM (Down syndrome cell adhesion molecule) as a candidate gene., Methods: A panel of 19 individuals with partial trisomy 21 was evaluated using quantitative Southern blot dosage analysis and fluorescence in situ hybridization (FISH) with subsets of 32 BACs spanning the region defined by D21S16 (21q11.2) through the telomere. These BACs span the molecular markers D21S55, ERG, ETS2, MX1/2, collagen XVIII and collagen VI A1/A2. Fourteen individuals are duplicated for the candidate region, of whom eight (57%) have the characteristic spectrum of DS-CHD., Results: Combining the results from these eight individuals suggests the candidate region for DS-CHD is demarcated by D21S3 (defined by ventricular septal defect), through PFKL (defined by tetralogy of Fallot)., Conclusions: These data suggest that the presence of three copies of gene(s) from the region is sufficient for the production of subsets of DS-CHD. This region does not include genes located near D21S55, previously proposed as a "DS critical region," or the genes encoding collagens VI and XVIII. Of the potential gene candidates in the narrowed DS-CHD region, DSCAM is notable in that it encodes a cell adhesion molecule, spans more than 840 kb of the candidate region, and is expressed in the heart during cardiac development. Given these properties, we propose DSCAM as a candidate for DS-CHD.
- Published
- 2001
- Full Text
- View/download PDF
45. Telokin expression is restricted to smooth muscle tissues during mouse development.
- Author
-
Herring BP, Lyons GE, Hoggatt AM, and Gallagher PJ
- Subjects
- Amino Acid Sequence physiology, Animals, Base Sequence physiology, Cloning, Molecular methods, DNA, Complementary genetics, Female, Genitalia, Female cytology, Genitalia, Female growth & development, Genitalia, Female metabolism, Genitalia, Male cytology, Genitalia, Male growth & development, Genitalia, Male metabolism, Male, Mice, Molecular Sequence Data, Muscle Development, Muscle Proteins metabolism, Muscle, Smooth cytology, Muscle, Smooth growth & development, Muscle, Smooth metabolism, Myosin-Light-Chain Kinase, Peptide Fragments, Peptides, RNA, Messenger genetics, RNA, Messenger metabolism, Gene Expression Regulation, Developmental physiology, Muscle Proteins genetics, Muscle, Smooth embryology
- Abstract
Telokin is a 17-kDa protein with an amino acid sequence that is identical to the COOH terminus of the 130-kDa myosin light chain kinase (MLCK). Telokin mRNA is transcribed from a second promoter, located within an intron, in the 3' region of the MLCK gene. In the current study, we show by in situ mRNA hybridization that telokin mRNA is restricted to the smooth muscle cell layers within adult smooth muscle tissues. In situ mRNA analysis of mouse embryos also revealed that telokin expression is restricted to smooth muscle tissues during embryonic development. Telokin mRNA expression was first detected in mouse gut at embryonic day 11.5; no telokin expression was detected in embryonic cardiac or skeletal muscle. Expression of telokin was also found to be regulated during postnatal development of the male and female reproductive tracts. In both uterus and vas deferens, telokin protein expression greatly increased between days 7 and 14 of postnatal development. The increase in telokin expression correlated with an increase in the expression of several other smooth muscle-restricted proteins, including smooth muscle myosin and alpha-actin.
- Published
- 2001
- Full Text
- View/download PDF
46. Down syndrome cell adhesion molecule is conserved in mouse and highly expressed in the adult mouse brain.
- Author
-
Barlow GM, Micales B, Lyons GE, and Korenberg JR
- Subjects
- Amino Acid Sequence, Animals, Brain cytology, Brain embryology, Brain growth & development, Cell Adhesion Molecules, Gene Expression Regulation, Developmental, Humans, In Situ Hybridization, Membrane Proteins, Molecular Sequence Data, Morphogenesis, Protein Structure, Tertiary, Proteins chemistry, RNA, Messenger genetics, RNA, Messenger metabolism, Sequence Homology, Amino Acid, Spinal Cord metabolism, Aging genetics, Brain metabolism, Conserved Sequence genetics, Down Syndrome genetics, Gene Expression Profiling, Mice genetics, Proteins genetics
- Abstract
Down Syndrome (DS) is a major cause of mental retardation and is associated with characteristic well-defined although subtle brain abnormalities, many of which arise after birth, with particular defects in the cortex, hippocampus and cerebellum. The neural cell adhesion molecule DSCAM (Down syndrome cell adhesion molecule) maps to 21q22.2-->q22.3, a region associated with DS mental retardation, and is expressed largely in the neurons of the central and peripheral nervous systems during development. In order to evaluate the contribution of DSCAM to postnatal morphogenetic and cognitive processes, we have analyzed the expression of the mouse DSCAM homolog, Dscam, in the adult mouse brain from 1 through 21 months of age. We have found that Dscam is widely expressed in the brain throughout adult life, with strongest levels in the cortex, the mitral and granular layers of the olfactory bulb, the granule cells of the dentate gyrus and the pyramidal cells of the CA1, CA2 and CA3 regions, the ventroposterior lateral nuclei of the thalamus, and in the Purkinje cells of the cerebellum. Dscam is also expressed ventrally in the adult spinal cord. Given the homology of DSCAM to cell adhesion molecules involved in development and synaptic plasticity, and its demonstrated role in axon guidance, we propose that DSCAM overexpression contributes not only to the structural defects seen in these regions of the DS brain, but also to the defects of learning and memory seen in adults with DS., (Copyright 2002 S. Karger AG, Basel)
- Published
- 2001
- Full Text
- View/download PDF
47. Transcriptional regulation of S100A1 and expression during mouse heart development.
- Author
-
Kiewitz R, Lyons GE, Schäfer BW, and Heizmann CW
- Subjects
- Animals, Calcium-Binding Proteins genetics, Cells, Cultured, Cloning, Molecular, Gene Expression Regulation, In Situ Hybridization, Luciferases genetics, Mice, Peptide Fragments genetics, Promoter Regions, Genetic, RNA, Messenger analysis, Restriction Mapping, S100 Proteins, Sequence Homology, Nucleic Acid, Species Specificity, Transcription, Genetic, Transfection, Calcium-Binding Proteins biosynthesis, Heart embryology
- Abstract
S100A1, a member of the large EF-hand family of Ca(2+)-binding proteins, is mainly expressed in the mammalian heart. To assess the underlying mechanisms for cell- and tissue-specific expression we isolated and characterized the mouse S100A1 gene. The gene displays a high degree of homology to the human and rat genes, especially in the exonic sequences. In its promoter region and the first intron, we identified regulatory elements characteristic for cardiac and slow skeletal muscle restricted genes. Transfection assays with luciferase constructs containing different parts of the S100A1 gene demonstrated the active expression in primary mouse cardiomyocytes and that its 5'-upstream region containing a putative cardiac enhancer showed a greatly increased activity. Furthermore, we investigated the expression of the S100A1 mRNA during embryonic mouse development, using in situ hybridization. S100A1 transcripts were first detected in the primitive heart at embryonic day (E) 8, with equal levels in the atrium and ventricle. During development up to E17.5 we detected a shift in the S100A1 expression pattern with lower levels in atrial and high levels in ventricular myocardium. The regulatory elements identified in the mouse S100A1 promoter correspond well with the observed expression pattern and suggest that S100A1 has an important function during heart muscle development.
- Published
- 2000
- Full Text
- View/download PDF
48. Jumonji, a nuclear protein that is necessary for normal heart development.
- Author
-
Lee Y, Song AJ, Baker R, Micales B, Conway SJ, and Lyons GE
- Subjects
- Animals, Biomarkers, Embryo, Mammalian physiology, Gene Expression, Genotype, Heart Defects, Congenital genetics, Heart Defects, Congenital metabolism, Heart Defects, Congenital pathology, Homozygote, Mice, Mice, Knockout genetics, Mice, Mutant Strains genetics, Mutation, Myocardium metabolism, Myocardium pathology, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nervous System metabolism, Nuclear Proteins physiology, Polycomb Repressive Complex 2, Tissue Distribution, Heart embryology, Nerve Tissue Proteins physiology
- Abstract
Jumonji (jmj) was cloned in a gene trap screen to identify and mutagenize genes important for heart development. To investigate the role of jmj in heart development, we generated mice homozygous for the jmj mutation. The jmj homozygous mouse embryos showed heart malformations, including ventricular septal defect, noncompaction of the ventricular wall, double-outlet right ventricle, and dilated atria. The jmj mutants died soon after birth, apparently as a result of respiratory insufficiency caused by rib and sternum defects in addition to the heart defects. In situ hybridization analyses suggested that cardiomyocytes were differentiated but developmental regulation of chamber-specific genes was defective in fetal hearts. Expression of jmj was detected in the myocardium, especially in the interventricular septum, ventricular wall, and outflow tract, which correlated well with the locations of defects observed in the hearts of mutant mice. Homozygous embryos failed to express the jmj transcript in all tissues except in the nervous system. Confocal microscopic examination using anti-JMJ antibodies indicated that the JMJ protein was localized in the nuclei of cells transfected with jmj. These data demonstrate that JMJ is a nuclear protein, which is essential for normal heart development and function.
- Published
- 2000
- Full Text
- View/download PDF
49. Gene trapping in embryonic stem cells in vitro to identify novel developmentally regulated genes in the mouse.
- Author
-
Lyons GE, Swanson BJ, Haendel MA, and Daniels J
- Subjects
- Animals, Embryo, Mammalian metabolism, In Vitro Techniques, Mice, Gene Expression Regulation, Developmental, Stem Cells metabolism
- Published
- 2000
- Full Text
- View/download PDF
50. Filamin isogene expression during mouse myogenesis.
- Author
-
Chiang W, Greaser ML, and Lyons GE
- Subjects
- Animals, Base Sequence, Embryonic and Fetal Development, Filamins, Mice, Molecular Sequence Data, Muscle, Skeletal embryology, Protein Isoforms biosynthesis, Protein Isoforms genetics, Contractile Proteins biosynthesis, Contractile Proteins genetics, Gene Expression Regulation, Developmental physiology, Microfilament Proteins biosynthesis, Microfilament Proteins genetics, Muscle, Skeletal physiology
- Abstract
The developmental pattern of filamin gene expression has been studied in mouse embryos by using in situ hybridization. The probes used were isoform specific, (35)S-labeled antisense complementary ribonucleic acids (cRNAs) to the 3; untranslated region (3; UTR) of muscle-specific and nonmuscle-specific filamin genes. Northern blot and in situ hybridization results showed that nonmuscle-specific filamin transcripts had a size of 9.5 kb and were expressed in all nonmuscle tissues. Labeling was most intense in tissues containing a substantial proportion of epithelial and smooth muscle cells. Muscle-specific filamin transcripts had a size of 10 kb and were expressed primarily in cardiac and skeletal muscle. The expression of muscle-specific filamin messenger ribonucleicacids (mRNAs) was detected in heart at 8.0 days after coitum, whereas that in the myotomes of somites was not detected until 10.5 days after coitum. The expression of muscle-specific filamin mRNAs in heart and in skeletal muscle continued through the subsequent days of myogenesis. The results showed that muscle-specific filamin gene transcripts are detected before the formation of myotubes in vivo. This is the first study of filamin gene expression at the early stages of skeletal muscle development. Dev Dyn 2000;217:99-108., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.