23 results on '"Hasneen K"'
Search Results
2. 766 Co-expression of oral epithelial regenerative factors Sox2 and Pitx1 induces wound activation signature in skin keratinocytes
- Author
-
Hope, E., primary, Overmiller, A., additional, Sawaya, A., additional, Uchiyama, A., additional, Nayak, S., additional, Hasneen, K., additional, Brooks, S., additional, and Morasso, M., additional
- Published
- 2022
- Full Text
- View/download PDF
3. 467 Crucial epigenetic modules in skin differentiation
- Author
-
Nayak, S., primary, Jiang, K., additional, Cross, M., additional, Hope, E., additional, Bajpai, D., additional, Worrell, S., additional, Hasneen, K., additional, Naz, F., additional, Brooks, S., additional, Dell'Orso, S., additional, and Morasso, M., additional
- Published
- 2022
- Full Text
- View/download PDF
4. 760 Cellular landscape of the skin is primed by the oral epithelial regenerative transcription factor Pitx1 to promote wound healing
- Author
-
Overmiller, A., primary, Uchiyama, A., additional, Hope, E., additional, Sawaya, A., additional, Nayak, S., additional, Hasneen, K., additional, Dell'Orso, S., additional, Brooks, S., additional, and Morasso, M., additional
- Published
- 2022
- Full Text
- View/download PDF
5. 662 Reprogramming of cutaneous keratinocytes by the regenerative transcription factor Pitx1 alters the cellular landscape of the skin and promotes wound healing
- Author
-
Overmiller, A., Uchiyama, A., Hope, E., O'Neill, C., Nayak, S., Hasneen, K., Dell'Orso, S., Brooks, S., and Morasso, M.I.
- Published
- 2024
- Full Text
- View/download PDF
6. 610 Deregulated immune signature orchestrated by FOXM1 impairs human diabetic wound healing
- Author
-
Sawaya, A., primary, Stone, R., additional, Brooks, S., additional, Pastar, I., additional, Jozic, I., additional, Hasneen, K., additional, O'Neill, K., additional, Mehdizadeh, S., additional, Head, C., additional, Strbo, N., additional, Morasso, M., additional, and Tomic-Canic, M., additional
- Published
- 2021
- Full Text
- View/download PDF
7. 611 Oral epithelial regenerative transcription factor Pitx1 reprograms keratinocytes to promote cutaneous wound healing
- Author
-
Overmiller, A., primary, Uchiyama, A., additional, Hope, E., additional, Grassini, D., additional, Sawaya, A., additional, Nayak, S., additional, Hasneen, K., additional, Chen, Y., additional, Brooks, S., additional, and Morasso, M., additional
- Published
- 2021
- Full Text
- View/download PDF
8. BMP4 Promotes Formation of Primitive Vascular Networks in Human Embryonic Stem Cell–Derived Embryoid Bodies.
- Author
-
Boyd, N. L., Dhara, S. K., Rekaya, R., Godbey, E. A., Hasneen, K., Rao, R. R., West III, F. D., Gerwe, B. A., and Stice, S. L.
- Published
- 2007
- Full Text
- View/download PDF
9. Reprogramming of epidermal keratinocytes by PITX1 transforms the cutaneous cellular landscape and promotes wound healing.
- Author
-
Overmiller AM, Uchiyama A, Hope ED, Nayak S, O'Neill CG, Hasneen K, Chen YW, Naz F, Dell'Orso S, Brooks SR, Jiang K, and Morasso MI
- Subjects
- Animals, Mice, Paired Box Transcription Factors genetics, Paired Box Transcription Factors metabolism, Cell Proliferation genetics, Cell Differentiation genetics, Cellular Reprogramming genetics, Cell Movement genetics, Epidermis metabolism, Mouth Mucosa metabolism, Mouth Mucosa cytology, Skin metabolism, Skin cytology, Keratinocytes metabolism, Wound Healing genetics
- Abstract
Cutaneous wound healing is a slow process that often terminates with permanent scarring while oral wounds, in contrast, regenerate after damage faster. Unique molecular networks in epidermal and oral epithelial keratinocytes contribute to the tissue-specific response to wounding, but key factors that establish those networks and how the keratinocytes interact with their cellular environment remain to be elucidated. The transcription factor PITX1 is highly expressed in the oral epithelium but is undetectable in cutaneous keratinocytes. To delineate if PITX1 contributes to oral keratinocyte identity, cell-cell interactions, and the improved wound healing capabilities, we ectopically expressed PITX1 in the epidermis of murine skin. Using comparative analysis of murine skin and oral (buccal) mucosa with single-cell RNA-Seq and spatial transcriptomics, we found that PITX1 expression enhances epidermal keratinocyte migration and proliferation and alters differentiation to a quasi-oral keratinocyte state. PITX1+ keratinocytes reprogrammed intercellular communication between skin-resident cells to mirror buccal tissue while stimulating the influx of neutrophils that establish a pro-inflammatory environment. Furthermore, PITX1+ skin healed significantly faster than control skin via increased keratinocyte activation and migration and a tunable inflammatory environment. These results illustrate that PITX1 programs oral keratinocyte identity and cellular interactions while revealing critical downstream networks that promote wound closure.
- Published
- 2024
- Full Text
- View/download PDF
10. SOX2-Dependent Wound Repair Signature Triggers Prohealing Outcome in Hyperglycemic Wounds.
- Author
-
O'Neill CG, Sawaya AP, Mehdizadeh S, Brooks SR, Hasneen K, Nayak S, Overmiller AM, and Morasso MI
- Published
- 2024
- Full Text
- View/download PDF
11. Chromatin Landscape Governing Murine Epidermal Differentiation.
- Author
-
Nayak S, Jiang K, Hope E, Cross M, Overmiller A, Naz F, Worrell S, Bajpai D, Hasneen K, Brooks SR, Dell'Orso S, and Morasso MI
- Subjects
- Mice, Animals, Cell Differentiation genetics, Epidermis metabolism, Epidermal Cells metabolism, Chromatin genetics, Chromatin metabolism, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Chromatin landscape and regulatory networks are determinants in lineage specification and differentiation. To define the temporospatial differentiation axis in murine epidermal cells in vivo, we generated datasets profiling expression dynamics (RNA sequencing), chromatin accessibility (assay for transposase-accessible chromatin using sequencing), architecture (Hi-C), and histone modifications (chromatin immunoprecipitation followed by sequencing) in the epidermis. We show that many differentially regulated genes are suppressed during the differentiation process, with superenhancers controlling differentiation-specific epigenomic changes. Our data shows the relevance of the Dlx/Klf/Grhl combinatorial regulatory network in maintaining correct temporospatial gene expression during epidermal differentiation. We determined differential open compartments, topologically associating domain score, and looping in the basal cell and suprabasal cell epidermal fractions, with the evolutionarily conserved epidermal differentiation complex region showing distinct suprabasal cell-specific topologically associating domain and loop formation that coincided with superenhancer sites. Overall, our study provides a global genome-wide resource of chromatin dynamics that define unrecognized regulatory networks and the epigenetic control of Dlx3-bound superenhancer elements during epidermal differentiation., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Loss of DLX3 tumor suppressive function promotes progression of SCC through EGFR-ERBB2 pathway.
- Author
-
Bajpai D, Mehdizadeh S, Uchiyama A, Inoue Y, Sawaya A, Overmiller A, Brooks SR, Hasneen K, Kellett M, Palazzo E, Motegi SI, Yuspa SH, Cataisson C, and Morasso MI
- Subjects
- Aged, Animals, Carcinogens toxicity, Carcinoma, Squamous Cell chemically induced, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Disease Models, Animal, ErbB Receptors genetics, ErbB Receptors metabolism, Female, Homeodomain Proteins metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Grading, Receptor, ErbB-2 genetics, Signal Transduction, Skin Neoplasms chemically induced, Skin Neoplasms genetics, Skin Neoplasms metabolism, Survival Rate, Tetradecanoylphorbol Acetate toxicity, Transcription Factors metabolism, 9,10-Dimethyl-1,2-benzanthracene toxicity, Carcinoma, Squamous Cell pathology, Homeodomain Proteins genetics, Receptor, ErbB-2 metabolism, Skin Neoplasms pathology, Transcription Factors genetics
- Abstract
Cutaneous squamous cell carcinoma (cSCC) ranks second in the frequency of all skin cancers. The balance between keratinocyte proliferation and differentiation is disrupted in the pathological development of cSCC. DLX3 is a homeobox transcription factor which plays pivotal roles in embryonic development and epidermal homeostasis. To investigate the impact of DLX3 expression on cSCC prognosis, we carried out clinicopathologic analysis of DLX3 expression which showed statistical correlation between tumors of higher pathologic grade and levels of DLX3 protein expression. Further, Kaplan-Meier survival curve analysis demonstrated that low DLX3 expression correlated with poor patient survival. To model the function of Dlx3 in skin tumorigenesis, a two-stage dimethylbenzanthracene (DMBA)/12-O-tetradecanoylphorbol 13-acetate (TPA) study was performed on mice genetically depleted of Dlx3 in skin epithelium (Dlx3cKO). Dlx3cKO mice developed significantly more tumors, with more rapid tumorigenesis compared to control mice. In Dlx3cKO mice treated only with DMBA, tumors developed after ~16 weeks suggesting that loss of Dlx3 has a tumor promoting effect. Whole transcriptome analysis of tumor and skin tissue from our mouse model revealed spontaneous activation of the EGFR-ERBB2 pathway in the absence of Dlx3. Together, our findings from human and mouse model system support a tumor suppressive function for DLX3 in skin and underscore the efficacy of therapeutic approaches that target EGFR-ERBB2 pathway.
- Published
- 2021
- Full Text
- View/download PDF
13. Deregulated immune cell recruitment orchestrated by FOXM1 impairs human diabetic wound healing.
- Author
-
Sawaya AP, Stone RC, Brooks SR, Pastar I, Jozic I, Hasneen K, O'Neill K, Mehdizadeh S, Head CR, Strbo N, Morasso MI, and Tomic-Canic M
- Subjects
- Adult, Aged, Animals, Cell Proliferation, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental immunology, Diabetic Foot pathology, Disease Models, Animal, Female, Forkhead Box Protein M1 antagonists & inhibitors, Forkhead Box Protein M1 metabolism, Humans, Inflammation genetics, Inflammation immunology, Male, Mice, Inbred Strains, Middle Aged, Mouth Mucosa physiology, Pyridines pharmacology, Thiophenes pharmacology, Transcriptome physiology, Wound Healing genetics, Diabetic Foot genetics, Diabetic Foot immunology, Forkhead Box Protein M1 immunology, Wound Healing immunology
- Abstract
Diabetic foot ulcers (DFUs) are a life-threatening disease that often result in lower limb amputations and a shortened lifespan. However, molecular mechanisms contributing to the pathogenesis of DFUs remain poorly understood. We use next-generation sequencing to generate a human dataset of pathogenic DFUs to compare to transcriptional profiles of human skin and oral acute wounds, oral as a model of "ideal" adult tissue repair due to accelerated closure without scarring. Here we identify major transcriptional networks deregulated in DFUs that result in decreased neutrophils and macrophages recruitment and overall poorly controlled inflammatory response. Transcription factors FOXM1 and STAT3, which function to activate and promote survival of immune cells, are inhibited in DFUs. Moreover, inhibition of FOXM1 in diabetic mouse models (STZ-induced and db/db) results in delayed wound healing and decreased neutrophil and macrophage recruitment in diabetic wounds in vivo. Our data underscore the role of a perturbed, ineffective inflammatory response as a major contributor to the pathogenesis of DFUs, which is facilitated by FOXM1-mediated deregulation of recruitment of neutrophils and macrophages, revealing a potential therapeutic strategy.
- Published
- 2020
- Full Text
- View/download PDF
14. Interferon lambda promotes immune dysregulation and tissue inflammation in TLR7-induced lupus.
- Author
-
Goel RR, Wang X, O'Neil LJ, Nakabo S, Hasneen K, Gupta S, Wigerblad G, Blanco LP, Kopp JB, Morasso MI, Kotenko SV, Yu ZX, Carmona-Rivera C, and Kaplan MJ
- Subjects
- Animals, B-Lymphocytes immunology, Cell Line, Gene Deletion, Humans, Imiquimod pharmacology, Inflammation immunology, Inflammation pathology, Interferon Inducers pharmacology, Interferon Type I physiology, Interferons pharmacology, Keratinocytes drug effects, Keratinocytes immunology, Keratinocytes pathology, Mesangial Cells drug effects, Mesangial Cells immunology, Mesangial Cells pathology, Mice, Inbred C57BL, Mice, Mutant Strains, Receptors, Interferon genetics, Signal Transduction, Toll-Like Receptor 7 agonists, Toll-Like Receptor 7 physiology, Interferon Lambda, Interferons physiology, Lupus Erythematosus, Systemic immunology, Lupus Erythematosus, Systemic pathology
- Abstract
Type III IFN lambdas (IFN-λ) have recently been described as important mediators of immune responses at barrier surfaces. However, their role in autoimmune diseases such as systemic lupus erythematosus (SLE), a condition characterized by aberrant type I IFN signaling, has not been determined. Here, we identify a nonredundant role for IFN-λ in immune dysregulation and tissue inflammation in a model of TLR7-induced lupus. IFN-λ protein is increased in murine lupus and IFN-λ receptor (Ifnlr1) deficiency significantly reduces immune cell activation and associated organ damage in the skin and kidneys without effects on autoantibody production. Single-cell RNA sequencing in mouse spleen and human peripheral blood revealed that only mouse neutrophils and human B cells are directly responsive to this cytokine. Rather, IFN-λ activates keratinocytes and mesangial cells to produce chemokines that induce immune cell recruitment and promote tissue inflammation. These data provide insights into the immunobiology of SLE and identify type III IFNs as important factors for tissue-specific pathology in this disease., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
15. SOX2 Epidermal Overexpression Promotes Cutaneous Wound Healing via Activation of EGFR/MEK/ERK Signaling Mediated by EGFR Ligands.
- Author
-
Uchiyama A, Nayak S, Graf R, Cross M, Hasneen K, Gutkind JS, Brooks SR, and Morasso MI
- Subjects
- Animals, Cell Proliferation genetics, Cells, Cultured, ErbB Receptors metabolism, Female, Heparin-binding EGF-like Growth Factor genetics, Keratinocytes metabolism, Ligands, Male, Mice, Models, Animal, Primary Cell Culture, RNA-Seq, SOXB1 Transcription Factors genetics, Signal Transduction genetics, Skin cytology, Skin metabolism, Up-Regulation, MAP Kinase Signaling System genetics, SOXB1 Transcription Factors metabolism, Skin injuries, Wound Healing genetics
- Abstract
Oral mucosa contains a unique transcriptional network that primes oral wounds for rapid resolution in humans. Our previous work identified genes that were consistently upregulated in the oral mucosa and demonstrated that induction of one of the identified genes, transcription factor SOX2, promoted cutaneous wound healing in mice. In this study, we investigated the molecular and cellular mechanisms by which SOX2 accelerates wound healing in skin. RNA-sequencing analysis showed that SOX2 induced a proliferative and wound-activated phenotype in skin keratinocytes prior to wounding. During wound healing, SOX2 induced proliferation of epithelial and connective tissue cells and promoted angiogenesis. Chromatin immunoprecipitation assay revealed that SOX2 directly regulates expression of EGFR ligands, resulting in activation of EGFR. In vitro, skin keratinocytes overexpressing SOX2 promoted cell migration via the EGFR/MEK/ERK pathway. We conclude that induction of SOX2 in skin keratinocytes accelerates cutaneous wound healing by promoting keratinocyte migration and proliferation, and enhancement of angiogenesis via upregulation of EGFR ligands and activation of EGFR/MEK/ERK pathway. Through the identification of putative cutaneous SOX2 targets, such as HBEGF, this study opens venues to determine clinical targets for treatment of skin wounds., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
16. Membrane proteomic signatures of karyotypically normal and abnormal human embryonic stem cell lines and derivatives.
- Author
-
Gerwe BA, Angel PM, West FD, Hasneen K, Young A, Orlando R, and Stice SL
- Subjects
- Cell Culture Techniques, Cell Differentiation, Cell Line, Chromatography, Reverse-Phase, Chromosome Duplication genetics, Electrophoresis, Polyacrylamide Gel, Embryonic Stem Cells cytology, Flow Cytometry, Gene Expression, Gene Expression Profiling, High-Throughput Screening Assays, Humans, Karyotyping, Mesenchymal Stem Cells cytology, Neural Stem Cells cytology, Proteomics methods, Spectroscopy, Fourier Transform Infrared, Trisomy genetics, Trypsin metabolism, Embryonic Stem Cells metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mesenchymal Stem Cells metabolism, Neural Stem Cells metabolism, Peptide Fragments analysis
- Abstract
Cultured human embryonic stem cells (hESCs) and derived derivatives contain heterogeneous cell populations with varying degrees of differentiation and karyotypic stability. The inability to isolate homogenous population presents a challenge toward cell-based applications and therapies. A proteomics approach was utilized to discover novel membrane proteins able to distinguish between the hESC lines BG01, WA09, and abBG02 (trisomy 12, 14, 17 and an extra copy of the X chromosome), along with WA09-derived human neural progenitor (hNP) cells. Membrane protein signatures were developed using sucrose-gradient isolation, 1-D gel electrophoresis followed by in-gel digestion and analysis by reverse phase chromatography coupled to ion trap-FT-ICR. At a ≤1.0% false discovery rate, 1918 proteins were identified; 775 were annotated as membrane proteins and 720 predicted to contain transmembrane spanning regions. Flow cytometry was used to validate cell surface expression of selected proteins. Junctional adhesion molecule 1 expression was shared by BG01, BG02 and abBG02 hESC lines. Dysferlin expression was specific to the WA09 hESC line and not the derived neural or mesenchymal progenitors. Ciliary neurotrophic factor receptor distinguished WA09-derived human neural progenitor cells from the parent hESC population, and WA09-derived mesenchymal progenitor cells. This study expands the current membrane protein data set for hESCs., (Copyright © 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2011
- Full Text
- View/download PDF
17. Differing lectin binding profiles among human embryonic stem cells and derivatives aid in the isolation of neural progenitor cells.
- Author
-
Dodla MC, Young A, Venable A, Hasneen K, Rao RR, Machacek DW, and Stice SL
- Subjects
- Cell Lineage, Cells, Cultured, Embryonic Stem Cells metabolism, Flow Cytometry, Humans, Immunohistochemistry, Neurons cytology, Neurons metabolism, Stem Cells metabolism, Cell Separation methods, Embryonic Stem Cells cytology, Lectins metabolism, Stem Cells cytology
- Abstract
Human embryonic stem cells (hESCs) and their differentiated progeny allow for investigation of important changes/events during normal embryonic development. Currently most of the research is focused on proteinacous changes occurring as a result of differentiation of stem cells and little is known about changes in cell surface glycosylation patterns. Identification of cell lineage specific glycans can help in understanding their role in maintenance, proliferation and differentiation. Furthermore, these glycans can serve as markers for isolation of homogenous populations of cells. Using a panel of eight biotinylated lectins, the glycan expression of hESCs, hESCs-derived human neural progenitors (hNP) cells, and hESCs-derived mesenchymal progenitor (hMP) cells was investigated. Our goal was to identify glycans that are unique for hNP cells and use the corresponding lectins for cell isolation. Flow cytometry and immunocytochemistry were used to determine expression and localization of glycans, respectively, in each cell type. These results show that the glycan expression changes upon differentiation of hESCs and is different for neural and mesenchymal lineage. For example, binding of PHA-L lectin is low in hESCs (14±4.4%) but significantly higher in differentiated hNP cells (99±0.4%) and hMP cells (90±3%). Three lectins: VVA, DBA and LTL have low binding in hESCs and hMP cells, but significantly higher binding in hNP cells. Finally, VVA lectin binding was used to isolate hNP cells from a mixed population of hESCs, hNP cells and hMP cells. This is the first report that compares glycan expression across these human stem cell lineages and identifies significant differences. Also, this is the first study that uses VVA lectin for isolation for human neural progenitor cells.
- Published
- 2011
- Full Text
- View/download PDF
18. Porcine induced pluripotent stem cells produce chimeric offspring.
- Author
-
West FD, Terlouw SL, Kwon DJ, Mumaw JL, Dhara SK, Hasneen K, Dobrinsky JR, and Stice SL
- Subjects
- Animal Structures cytology, Animal Structures metabolism, Animals, Animals, Newborn abnormalities, Animals, Newborn metabolism, Basic Helix-Loop-Helix Transcription Factors genetics, Blastocyst cytology, Cell Differentiation genetics, Chimera abnormalities, Chimera metabolism, Embryoid Bodies cytology, Fetal Proteins genetics, Fetus cytology, Fetus metabolism, Gene Expression genetics, Homeodomain Proteins genetics, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells transplantation, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors genetics, Mesenchymal Stem Cells cytology, Nanog Homeobox Protein, Nerve Tissue Proteins genetics, Octamer Transcription Factor-3 genetics, Octamer Transcription Factor-3 metabolism, Proto-Oncogene Proteins c-myc genetics, RNA-Binding Proteins genetics, SOXB1 Transcription Factors genetics, SOXB1 Transcription Factors metabolism, T-Box Domain Proteins genetics, Transduction, Genetic, alpha-Fetoproteins genetics, Brachyury Protein, Chimera embryology, Induced Pluripotent Stem Cells cytology, Sus scrofa
- Abstract
Ethical and moral issues rule out the use of human induced pluripotent stem cells (iPSCs) in chimera studies that would determine the full extent of their reprogrammed state, instead relying on less rigorous assays such as teratoma formation and differentiated cell types. To date, only mouse iPSC lines are known to be truly pluripotent. However, initial mouse iPSC lines failed to form chimeric offspring, but did generate teratomas and differentiated embryoid bodies, and thus these specific iPSC lines were not completely reprogrammed or truly pluripotent. Therefore, there is a need to address whether the reprogramming factors and process used eventually to generate chimeric mice are universal and sufficient to generate reprogrammed iPSC that contribute to chimeric offspring in additional species. Here we show that porcine mesenchymal stem cells transduced with 6 human reprogramming factors (POU5F1, SOX2, NANOG, KLF4, LIN28, and C-MYC) injected into preimplantation-stage embryos contributed to multiple tissue types spanning all 3 germ layers in 8 of 10 fetuses. The chimerism rate was high, 85.3% or 29 of 34 live offspring were chimeras based on skin and tail biopsies harvested from 2- to 5-day-old pigs. The creation of pluripotent porcine iPSCs capable of generating chimeric offspring introduces numerous opportunities to study the facets significantly affecting cell therapies, genetic engineering, and other aspects of stem cell and developmental biology.
- Published
- 2010
- Full Text
- View/download PDF
19. Effects of topography on the functional development of human neural progenitor cells.
- Author
-
Wu ZZ, Kisaalita WS, Wang L, Zachman AL, Zhao Y, Hasneen K, Machacek D, and Stice SL
- Subjects
- Calcium metabolism, Calcium Channels physiology, Cell Culture Techniques, Cell Line, Humans, Microscopy, Electron, Scanning, Microspheres, Neuromuscular Depolarizing Agents metabolism, Neurons cytology, Polystyrenes, Potassium metabolism, Stem Cells cytology, Neurons physiology, Stem Cells physiology
- Abstract
We have fabricated a topographical substrate with a packed polystyrene bead array for the development of cell-based assay systems targeting voltage-gated calcium channels (VGCCs). Human neural progenitor cells (H945RB.3) cultured on both flat and topographical substrates were analyzed in terms of morphological spreading, neuronal commitment, resting membrane potential (V(m)) establishment and VGCC function development. We found, by SEM imaging, that arrayed substrates, formed with both sub-micrometer (of 0.51 microm in mean diameter) and micrometer (of 1.98 microm in mean diameter) beads, were capable of promoting the spreading of the progenitor cells as compared with the flat polystyrene surfaces. With the micrometer beads, it was found that arrayed substrates facilitated the neural progenitor cells' maintenance of less negative V(m) values upon differentiation with bFGF starvation, which favored predominant neuronal commitment. Almost all the progenitor cells were responsive to 50 mM K(+) depolarization with an increase in [Ca(2+)](i) either before or upon differentiation, suggesting the expression of functional VGCCs. Compared to the flat polystyrene surfaces, microbead arrayed substrates facilitated the development of higher VGCC responsiveness by the progenitor cells upon differentiation. The enhancement of both VGCC responsiveness and cell spreading by arrays of micrometer beads was most significant on day 14 into differentiation, which was the latest time point of measurement in this study. This study thus rationalized the possibility for future substrate topography engineering to manipulate ion channel function and to meet the challenge of low VGCC responsiveness found in early drug discovery.
- Published
- 2010
- Full Text
- View/download PDF
20. Plasma from systemic lupus patients compromises cholesterol homeostasis: a potential mechanism linking autoimmunity to atherosclerotic cardiovascular disease.
- Author
-
Reiss AB, Anwar K, Merrill JT, Chan ES, Awadallah NW, Cronstein BN, Michael Belmont H, Belilos E, Rosenblum G, Belostocki K, Bonetti L, Hasneen K, and Carsons SE
- Subjects
- Adolescent, Adult, Atherosclerosis blood, Case-Control Studies, Cells, Cultured, Cholestanetriol 26-Monooxygenase genetics, Down-Regulation, Endothelial Cells immunology, Female, Foam Cells enzymology, Gene Expression Regulation, Enzymologic, Homeostasis, Humans, Interferon-gamma antagonists & inhibitors, Interferon-gamma immunology, Lipoproteins, LDL metabolism, Lupus Erythematosus, Systemic complications, Lupus Erythematosus, Systemic immunology, Monocytes immunology, RNA, Messenger metabolism, Receptors, Interferon antagonists & inhibitors, Receptors, Interferon immunology, Risk Factors, Young Adult, Interferon gamma Receptor, Atherosclerosis immunology, Autoimmunity, Cholestanetriol 26-Monooxygenase metabolism, Cholesterol metabolism, Endothelial Cells enzymology, Lupus Erythematosus, Systemic blood, Monocytes enzymology
- Abstract
Atherosclerotic cardiovascular disease (ASCVD) contributes to morbidity and mortality in systemic lupus erythematosus (SLE). Immunologic derangements may disrupt cholesterol balance in vessel wall monocytes/macrophages and endothelium. We determined whether lupus plasma impacts expression of cholesterol 27-hydroxylase, an anti-atherogenic cholesterol-degrading enzyme that promotes cellular cholesterol efflux, in THP-1 human monocytes and primary human aortic endothelial cells (HAEC). THP-1 monocytes and HAEC were incubated in medium containing SLE patient plasma or apparently healthy control human plasma (CHP). SLE plasma decreased 27-hydroxylase message in THP-1 monocytes by 47 +/- 8% (p < 0.008) and in HAEC by 51 +/- 5.5% (n = 5, p < 0.001). THP-1 macrophages were incubated in 25% lupus plasma or CHP and cholesterol-loaded (50 microg ml(-1) acetylated low density lipoprotein). Lupus plasma more than doubled macrophage foam cell transformation (74 +/- 3% vs. 35 +/- 3% for CHP, n = 3, p < 0.001). Impaired cholesterol homeostasis in SLE provides further evidence of immune involvement in atherogenesis. Strategies to inhibit or reverse arterial cholesterol accumulation may benefit SLE patients.
- Published
- 2010
- Full Text
- View/download PDF
21. Genetic manipulation of neural progenitors derived from human embryonic stem cells.
- Author
-
Dhara SK, Gerwe BA, Majumder A, Dodla MC, Boyd NL, Machacek DW, Hasneen K, and Stice SL
- Subjects
- Cell Differentiation genetics, Cell Proliferation, Cell Survival genetics, Cells, Cultured, Humans, Embryonic Stem Cells cytology, Embryonic Stem Cells physiology, Genetic Enhancement methods, Neurons cytology, Neurons physiology
- Abstract
Human embryonic stem cell-derived neural progenitors (NP) present an important tool for understanding human development and disease. Optimal utilization of NP cells, however, requires an enhanced ability to monitor these cells in vitro and in vivo. Here we report production of the first genetically modified self-renewing human embryonic stem cell-derived NP cells that express fluorescent proteins under constitutive as well as lineage-specific promoters, enabling tracking and monitoring of cell fate. Nucleofection, transfection, and lentiviral transduction were compared for optimal gene delivery to NP cells. Transduction was most efficient in terms of transgene expression (37%), cell viability (39%), and long-term reporter expression (>3 months). Further, the constitutive gene promoters, cytomegalovirus, elongation factor 1alpha, and ubiquitin-C, exhibited comparable silencing (20-30%) in NP cells over a 2-month period, suggesting their suitability for long-term reporter expression studies. Transduced NP cells maintained their progenitor state and differentiation potential, as demonstrated by expression of endogenous NP markers and neuronal markers after differentiation. We also detected reporter expression in astrocytes generated from NP cells transduced with an astrocyte-specific gene promoter, glial fibrillary acidic protein, demonstrating the usefulness of this approach. The genetically manipulated NP cells described here offer great potential for live cell-tracking experiments, and a similar approach can as well be used for expression of proteins other than reporters.
- Published
- 2009
- Full Text
- View/download PDF
22. Human neural progenitor cells derived from embryonic stem cells in feeder-free cultures.
- Author
-
Dhara SK, Hasneen K, Machacek DW, Boyd NL, Rao RR, and Stice SL
- Subjects
- Antigens, Differentiation biosynthesis, Antigens, Differentiation genetics, Astrocytes metabolism, Cell Differentiation drug effects, Cell Division drug effects, Culture Media chemistry, Culture Media, Conditioned pharmacology, Culture Media, Serum-Free pharmacology, Embryonic Stem Cells drug effects, Gene Expression Profiling, Gene Expression Regulation, Developmental drug effects, Humans, Laminin, Nerve Tissue Proteins biosynthesis, Nerve Tissue Proteins genetics, Neurons metabolism, Oligodendroglia metabolism, Pluripotent Stem Cells cytology, Pluripotent Stem Cells drug effects, RNA, Messenger biosynthesis, RNA, Messenger genetics, Reproducibility of Results, Astrocytes cytology, Cell Culture Techniques methods, Culture Media pharmacology, Embryonic Stem Cells cytology, Neurons cytology, Oligodendroglia cytology
- Abstract
Derivation of human neural progenitors (hNP) from human embryonic stem (hES) cells in culture has been reported with the use of feeder cells or conditioned media. This introduces undefined components into the system, limiting the ability to precisely investigate the requirement for factors that control the process. Also, the use of feeder cells of non-human origin introduces the potential for zoonotic transmission, limiting its clinical usefulness. Here we report a feeder-free system to produce hNP from hES cells and test the effects of various media components involved in the process. Five protocols using defined media components were compared for efficiency of hNP generation. Based on this analysis, we discuss the role of basic fibroblast growth factor (FGF2), N2 supplement, non-essential amino acids (NEAA), and knock-out serum replacement (KSR) on the process of hNP generation. All protocols led to down-regulation of Oct4/POU5F1 expression (from 90.5% to <3%), and up-regulation of neural progenitor markers to varying degrees. Media with N2 but not KSR and NEAA produced cultures with significantly higher (p<0.05) expression of the neural progenitor marker Musashi 1 (MSI1). Approximately 89% of these cells were Nestin (NES)+ after 3 weeks, but they did not proliferate. In contrast, differentiation media supplemented with KSR and NEAA produced fewer NES+ (75%) cells, but these cells were proliferative, and by five passages the culture consisted of >97% NES+ cells. This suggests that KSR and NEAA supplements did not enhance early differentiation but did promote proliferating of hNP cell cultures. This resulted in an efficient, robust, repeatable differentiation system suitable for generating large populations of hNP cells. This will facilitate further study of molecular and biochemical mechanisms in early human neural differentiation and potentially produce uniform neuronal cells for therapeutic uses without concern of zoonotic transmission from feeder layers.
- Published
- 2008
- Full Text
- View/download PDF
23. Interferon-gamma impedes reverse cholesterol transport and promotes foam cell transformation in THP-1 human monocytes/macrophages.
- Author
-
Reiss AB, Patel CA, Rahman MM, Chan ES, Hasneen K, Montesinos MC, Trachman JD, and Cronstein BN
- Subjects
- Antibodies immunology, Arteriosclerosis etiology, Biological Transport drug effects, Cell Differentiation, Cell Line, Cholestanetriol 26-Monooxygenase, Cholesterol, LDL metabolism, Down-Regulation genetics, Foam Cells chemistry, Foam Cells drug effects, Humans, Interferon-gamma immunology, Interferon-gamma metabolism, Steroid Hydroxylases genetics, Cholesterol metabolism, Foam Cells metabolism, Interferon-gamma pharmacology, Macrophages cytology, Monocytes cytology, Steroid Hydroxylases metabolism
- Abstract
Background: Cholesterol 27-hydroxylase, an enzyme expressed at high levels by human monocytes/macrophages, provides a first line of defense against the development of atherosclerosis. Prior studies have suggested that the cytokine interferon-gamma (IFN-gamma) promotes atherosclerosis. We therefore examined the effect of IFN-g on macrophage foam cell formation and on expression of the anti-atherogenic 27-hydroxylase in THP-1 human monocytes/macrophages., Material/methods: THP-1 monocytes and acetylated LDL-treated THP-1 macrophages were incubated in the presence or absence of IFN-gamma (500 U/ml) with or without the addition of IFN- gamma receptor blocking or neutralizing antibody. Foam cell formation was quantified based on percentage of macrophages harboring oil red O-stained globules. Cellular mRNA and protein were isolated. 27-Hydroxylase message was measured by RT-PCR and 27-hydroxylase protein by immunoblot., Results: IFN-gamma -treated THP-1 macrophages exhibit increased foam cell transformation compared to untreated cells under cholesterol loading conditions. IFN-gamma-promoted foam cell formation is abolished by pre-treatment with either IFN-gamma neutralizing or IFN-gamma receptor blocking antibody. IFN-gamma diminishes cholesterol 27-hydroxylase expression in THP-1, and this IFN-gamma -induced downregulation is prevented by pre-treating the cultured cells with either IFN-gamma neutralizing or IFN-gamma receptor blocking antibody., Conclusions: Imbalances in cellular cholesterol flux within macrophages lead to formation of lipid-laden foam cells, a critical step in the pathogenesis of atherosclerosis. We have demonstrated that IFN-gamma, acting through the IFN-gamma receptor, decreases expression of 27-hydroxylase and increases propensity to foam cell formation in the cell line THP-1. These observations suggest that one mechanism by which IFN-g promotes atherosclerosis may involve affecting expression of cholesterol 27-hydroxylase, a cholesterol homeostatic protein.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.