Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
STAR Protoc ; 4(3): 102552, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37651236

ABSTRACT

Nuclear pore complexes are pathways for nuclear-cytoplasmic communication that participate in chromatin organization. Here, we present a protocol to image and quantify the number of nuclear pore complexes in cells. We describe steps for cell plating and culture, immunofluorescence detection, and confocal microscopy visualization of nuclear pore complexes. We then detail quantification and 3D data analysis. This protocol utilizes digital thresholding under human supervision for quantification of nuclear pore complexes. For complete details on the use and execution of this protocol, please refer to Han et al.1.


Subject(s)
Data Analysis , Nuclear Pore , Humans , Cytoplasm , Cytosol , Microscopy, Confocal
2.
Dev Cell ; 57(20): 2397-2411.e9, 2022 10 24.
Article in English | MEDLINE | ID: mdl-36283391

ABSTRACT

Nuclear pores are essential for nuclear-cytoplasmic transport. Whether and how cells change nuclear pores to alter nuclear transport and cellular function is unknown. Here, we show that rat heart muscle cells (cardiomyocytes) undergo a 63% decrease in nuclear pore numbers during maturation, and this changes their responses to extracellular signals. The maturation-associated decline in nuclear pore numbers is associated with lower nuclear import of signaling proteins such as mitogen-activated protein kinase (MAPK). Experimental reduction of nuclear pore numbers decreased nuclear import of signaling proteins, resulting in decreased expression of immediate-early genes. In a mouse model of high blood pressure, reduction of nuclear pore numbers improved adverse heart remodeling and reduced progression to lethal heart failure. The decrease in nuclear pore numbers in cardiomyocyte maturation and resulting functional changes demonstrate how terminally differentiated cells permanently alter their handling of information flux across the nuclear envelope and, with that, their behavior.


Subject(s)
Nuclear Envelope , Nuclear Pore , Mice , Rats , Animals , Nuclear Pore/metabolism , Active Transport, Cell Nucleus , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism
3.
Methods Mol Biol ; 2158: 125-139, 2021.
Article in English | MEDLINE | ID: mdl-32857370

ABSTRACT

Failure to regenerate myocardium after injury is a major cause of mortality and morbidity in humans. Direct differentiation of human induced pluripotent stem cells (iPSCs) into cardiomyocytes provides an invaluable resource to pursue cardiac regeneration based on cellular transplantation. Beyond the potential for clinical therapies, iPSC technology also enables the generation of cardiomyocytes to recapitulate patient-specific phenotypes, thus presenting a powerful in vitro cell-based model to understand disease pathology and guide precision medicine. Here, we describe protocols for reprogramming of human dermal fibroblasts and blood cells into iPSCs using the non-integrative Sendai virus system and for the monolayer differentiation of iPSCs to cardiomyocytes using chemically defined media.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Dermis/cytology , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Regeneration , Humans
4.
Dev Cell ; 53(1): 42-59.e11, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32109383

ABSTRACT

Heart regeneration requires cardiomyocyte proliferation. It is thought that formation of polyploid nuclei establishes a barrier for cardiomyocyte proliferation, but the mechanisms are largely unknown. Here, we show that the nuclear lamina filament Lamin B2 (Lmnb2), whose expression decreases in mice after birth, is essential for nuclear envelope breakdown prior to progression to metaphase and subsequent division. Inactivating Lmnb2 decreased metaphase progression, which led to formation of polyploid cardiomyocyte nuclei in neonatal mice, which, in turn, decreased myocardial regeneration. Increasing Lmnb2 expression promoted cardiomyocyte M-phase progression and cytokinesis and improved indicators of myocardial regeneration in neonatal mice. Inactivating LMNB2 in human iPS cell-derived cardiomyocytes reduced karyokinesis and increased formation of polyploid nuclei. In primary cardiomyocytes from human infants with heart disease, modifying LMNB2 expression correspondingly altered metaphase progression and ploidy of daughter nuclei. In conclusion, Lmnb2 expression is essential for karyokinesis in mammalian cardiomyocytes and heart regeneration.


Subject(s)
Heart/physiology , Lamin Type B/metabolism , Myocytes, Cardiac/metabolism , Regeneration/physiology , Animals , Cell Nucleus/metabolism , Cell Nucleus Division/physiology , Cell Proliferation/physiology , Cells, Cultured , Induced Pluripotent Stem Cells/cytology , Mice , Wound Healing/physiology
6.
Cardiovasc Res ; 104(2): 258-69, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25209314

ABSTRACT

AIMS: Familial hypertrophic cardiomyopathy (HCM) is one the most common heart disorders, with gene mutations in the cardiac sarcomere. Studying HCM with patient-specific induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs) would benefit the understanding of HCM mechanism, as well as the development of personalized therapeutic strategies. METHODS AND RESULTS: To investigate the molecular mechanism underlying the abnormal CM functions in HCM, we derived iPSCs from an HCM patient with a single missense mutation (Arginine442Glycine) in the MYH7 gene. CMs were next enriched from HCM and healthy iPSCs, followed with whole transcriptome sequencing and pathway enrichment analysis. A widespread increase of genes responsible for 'Cell Proliferation' was observed in HCM iPSC-CMs when compared with control iPSC-CMs. Additionally, HCM iPSC-CMs exhibited disorganized sarcomeres and electrophysiological irregularities. Furthermore, disease phenotypes of HCM iPSC-CMs were attenuated with pharmaceutical treatments. CONCLUSION: Overall, this study explored the possible patient-specific and mutation-specific disease mechanism of HCM, and demonstrates the potential of using HCM iPSC-CMs for future development of therapeutic strategies. Additionally, the whole methodology established in this study could be utilized to study mechanisms of other human-inherited heart diseases.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/genetics , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Adult , Animals , Calcium Signaling/genetics , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/pathology , Case-Control Studies , Cell Proliferation/genetics , Cell Separation/methods , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Mice, Inbred NOD , Mice, SCID , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Myosin Heavy Chains/genetics , Phenotype , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Transcriptome
7.
PLoS One ; 9(6): e100417, 2014.
Article in English | MEDLINE | ID: mdl-24949734

ABSTRACT

Hepatocyte transplantation is considered to be a promising therapy for patients with liver diseases. Induced pluripotent stem cells (iPSCs) provide an unlimited source for the generation of functional hepatocytes. In this study, we generated iPSCs from porcine ear fibroblasts (PEFs) by overexpressing Sox2, Klf4, Oct4, and c-Myc (SKOM), and developed a novel strategy for the efficient differentiation of hepatocyte-like cells from porcine iPSCs by following the processes of early liver development. The differentiated cells displayed the phenotypes of hepatocytes, exhibited classic hepatocyte-associated bio-functions, such as LDL uptake, glycogen storage and urea secretion, as well as possessed the metabolic activities of cytochrome P-450 (CYP) 3A and 2C. Furthermore, we compared the hepatocyte differentiation efficacy of our protocol with another published method, and the results demonstrated that our differentiation strategy could significantly improve the generation of morphological and functional hepatocyte-like cells from porcine iPSCs. In conclusion, this study establishes an efficient method for in vitro generation of functional hepatocytes from porcine iPSCs, which could represent a promising cell source for preclinical testing of cell-based therapeutics for liver failure and for pharmacological applications.


Subject(s)
Cellular Reprogramming , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Swine , Animals , Fibroblasts/cytology , Gene Expression Profiling , Genetic Vectors/genetics , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Lentivirus/genetics , Liver/cytology , Liver/growth & development , Male , Organ Specificity
8.
PLoS One ; 8(2): e56831, 2013.
Article in English | MEDLINE | ID: mdl-23437251

ABSTRACT

Corneal transparency depends on a unique extracellular matrix secreted by stromal keratocytes, mesenchymal cells of neural crest lineage. Derivation of keratocytes from human embryonic stem (hES) cells could elucidate the keratocyte developmental pathway and open a potential for cell-based therapy for corneal blindness. This study seeks to identify conditions inducing differentiation of pluripotent hES cells to the keratocyte lineage. Neural differentiation of hES cell line WA01(H1) was induced by co-culture with mouse PA6 fibroblasts. After 6 days of co-culture, hES cells expressing cell-surface NGFR protein (CD271, p75NTR) were isolated by immunoaffinity adsorption, and cultured as a monolayer for one week. Keratocyte phenotype was induced by substratum-independent pellet culture in serum-free medium containing ascorbate. Gene expression, examined by quantitative RT-PCR, found hES cells co-cultured with PA6 cells for 6 days to upregulate expression of neural crest genes including NGFR, SNAI1, NTRK3, SOX9, and MSX1. Isolated NGFR-expressing cells were free of PA6 feeder cells. After expansion as a monolayer, mRNAs typifying adult stromal stem cells were detected, including BMI1, KIT, NES, NOTCH1, and SIX2. When these cells were cultured as substratum-free pellets keratocyte markers AQP1, B3GNT7, PTDGS, and ALDH3A1 were upregulated. mRNA for keratocan (KERA), a cornea-specific proteoglycan, was upregulated more than 10,000 fold. Culture medium from pellets contained high molecular weight keratocan modified with keratan sulfate, a unique molecular component of corneal stroma. These results show hES cells can be induced to differentiate into keratocytes in vitro. Pluripotent stem cells, therefore, may provide a renewable source of material for development of treatment of corneal stromal opacities.


Subject(s)
Cell Differentiation , Corneal Keratocytes/cytology , Embryonic Stem Cells/cytology , Animals , Cell Line , Coculture Techniques , Corneal Keratocytes/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Immunophenotyping , Mice , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Organ Specificity/genetics , Phenotype , Proteoglycans/metabolism , Receptor, Nerve Growth Factor/metabolism , Stromal Cells/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Carbohydrate Sulfotransferases
9.
PLoS One ; 7(11): e49700, 2012.
Article in English | MEDLINE | ID: mdl-23209593

ABSTRACT

Human cytomegalovirus (HCMV) infection is one of the leading prenatal causes of congenital mental retardation and deformities world-wide. Access to cultured human neuronal lineages, necessary to understand the species specific pathogenic effects of HCMV, has been limited by difficulties in sustaining primary human neuronal cultures. Human induced pluripotent stem (iPS) cells now provide an opportunity for such research. We derived iPS cells from human adult fibroblasts and induced neural lineages to investigate their susceptibility to infection with HCMV strain Ad169. Analysis of iPS cells, iPS-derived neural stem cells (NSCs), neural progenitor cells (NPCs) and neurons suggests that (i) iPS cells are not permissive to HCMV infection, i.e., they do not permit a full viral replication cycle; (ii) Neural stem cells have impaired differentiation when infected by HCMV; (iii) NPCs are fully permissive for HCMV infection; altered expression of genes related to neural metabolism or neuronal differentiation is also observed; (iv) most iPS-derived neurons are not permissive to HCMV infection; and (v) infected neurons have impaired calcium influx in response to glutamate.


Subject(s)
Cytomegalovirus/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/virology , Neurons/cytology , Neurons/virology , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/virology , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Neurons/metabolism , Viral Tropism
10.
Stem Cell Res ; 7(1): 28-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543277

ABSTRACT

Unequivocal evidence for pluripotency in which embryonic stem cells contribute to chimeric offspring has yet to be demonstrated in human or nonhuman primates (NHPs). Here, rhesus and baboons ESCs were investigated in interspecific mouse chimera generated by aggregation or blastocyst injection. Aggregation chimera produced mouse blastocysts with GFP-nhpESCs at the inner cell mass (ICM), and embryo transfers (ETs) generated dimly-fluorescencing abnormal fetuses. Direct injection of GFP-nhpESCs into blastocysts produced normal non-GFP-fluorescencing fetuses. Injected chimera showed >70% loss of GFP-nhpESCs after 21 h culture. Outgrowths of all chimeric blastocysts established distinct but separate mouse- and NHP-ESC colonies. Extensive endogenous autofluorescence compromised anti-GFP detection and PCR analysis did not detect nhpESCs in fetuses. NhpESCs localize to the ICM in chimera and generate pregnancies. Because primate ESCs do not engraft post-implantation, and also because endogenous autofluorescence results in misleading positive signals, interspecific chimera assays for pluripotency with primate stem cells is unreliable with the currently available ESCs. Testing primate ESCs reprogrammed into even more naïve states in these inter-specific chimera assays will be an important future endeavor.


Subject(s)
Chimera/embryology , Embryonic Stem Cells/cytology , Haplorhini/embryology , Animals , Embryo Implantation , Embryo Transfer , Embryo, Mammalian/cytology , Female , Humans , Male , Mice , Mice, Inbred ICR , Pregnancy
11.
Stem Cell Res ; 5(3): 201-11, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20864427

ABSTRACT

Teratoma formation in xenografts is a sufficiently stringent pluripotency assay for stem cells. However, little is known about the composition and spatial relationships of tissues within teratomas that may provide clues about development and platforms for studying organ development. Additionally, teratoma formation and analysis lack standards for reporting as assays of pluripotency. Three of 27 total teratomas derived from pedigreed primate embryonic stem cells underwent quantitative three-dimensional high-resolution magnetic resonance microscopy (MRM). Teratomas were subsequently serially sectioned and tissue types identified, semiquantitated, and correlated with MRM images. All teratomas demonstrated tissue derivatives from the three germ layers and approximately 23 different tissue types were identified. Certain tissue groups attempted to form organs more frequently (e.g., trachea/bronchi, small intestine). MRM discriminated some tissues readily (e.g., bone, adipose, cartilage) while other tissue types with like MR intensities could not be distinguished. Semiquantitative histopathological analysis of teratomas demonstrates the ability to delineate multiple tissues as derived from ectoderm, mesoderm, or endoderm and to use this information for comparison to other teratomas. MRM provides rapid quantitative imaging of intact teratomas that complements histology and identifies sites of interest for additional biological studies.


Subject(s)
Embryonic Stem Cells/pathology , Teratoma/pathology , Animals , Microscopy/instrumentation , Microscopy/methods , Primates
12.
Stem Cell Res ; 4(1): 25-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19854689

ABSTRACT

While human embryonic stem cells (hESCs) are predisposed toward chromosomal aneploidities on 12, 17, 20, and X, rendering them susceptible to transformation, the specific genes expressed are not yet known. Here, by identifying the genes overexpressed in pluripotent rhesus ESCs (nhpESCs) and comparing them both to their genetically identical differentiated progeny (teratoma fibroblasts) and to genetically related differentiated parental cells (parental skin fibroblasts from whom gametes were used for ESC derivation), we find that some of those overexpressed genes in nhpESCs cluster preferentially on rhesus chromosomes 16, 19, 20, and X, homologues of human chromosomes 17, 19, 16, and X, respectively. Differentiated parental skin fibroblasts display gene expression profiles closer to nhpESC profiles than to teratoma cells, which are genetically identical to the pluripotent nhpESCs. Twenty over- and underexpressed pluripotency modulators, some implicated in neurogenesis, have been identified. The overexpression of some of these genes discovered using pedigreed nhpESCs derived from prime embryos generated by fertile primates, which is impossible to perform with the anonymously donated clinically discarded embryos from which hESCs are derived, independently confirms the importance of chromosome 17 and X regions in pluripotency and suggests specific candidates for targeting differentiation and transformation decisions.


Subject(s)
Chromosomes, Human , Embryonic Stem Cells/metabolism , Gene Expression , Macaca mulatta/genetics , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Cell Line , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 19 , Chromosomes, Human, X , Embryonic Stem Cells/cytology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Pluripotent Stem Cells/cytology , Teratoma/genetics , Teratoma/pathology
13.
Stem Cell Res ; 2(3): 178-87, 2009 May.
Article in English | MEDLINE | ID: mdl-19393591

ABSTRACT

Here we have developed protocols using the baboon as a complementary alternative Old World Primate to rhesus and other macaques which have severe limitations in their availability. Baboons are not limited as research resources, they are evolutionarily closer to humans, and the multiple generations of pedigreed colonies which display complex human disease phenotypes all support their further optimization as an invaluable primate model. Since neither baboon-assisted reproductive technologies nor baboon embryonic stem cells (ESCs) have been reported, here we describe the first derivations and characterization of baboon ESC lines from IVF-generated blastocysts. Two ESCs lines (BabESC-4 and BabESC-15) display ESC morphology, express pluripotency markers (Oct-4, hTert, Nanog, Sox-2, Rex-1, TRA1-60, TRA1-81), and maintain stable euploid female karyotypes with parentage confirmed independently. They have been grown continuously for >430 and 290 days, respectively. Teratomas from both lines have all three germ layers. Availabilities of these BabESCs represent another important resource for stem cell biologists.


Subject(s)
Cell Line , Embryonic Stem Cells/cytology , Models, Biological , Animals , Biomarkers/metabolism , Blastomeres/cytology , Cell Differentiation , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Karyotyping , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Papio , Primates , Regenerative Medicine , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Telomerase/genetics , Telomerase/metabolism
14.
Stem Cells ; 25(11): 2695-2704, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17641389

ABSTRACT

Human embryonic stem cells (hESCs) hold great biomedical promise, but experiments comparing them produce heterogeneous results, raising concerns regarding their reliability and utility, although these variations may result from their disparate and anonymous origins. To determine whether primate ESCs have intrinsic biological limitations compared with mouse ESCs, we examined expression profiles and pluripotency of newly established nonhuman primate ESC (nhpESCs). Ten pedigreed nhpESC lines, seven full siblings (fraternal quadruplets and fraternal triplets), and nine half siblings were derived from 41 rhesus embryos; derivation success correlated with embryo quality. Each line has been growing continuously for approximately 1 year with stable diploid karyotype (except for one stable trisomy) and expresses in vitro pluripotency markers, and eight have already formed teratomas. Unlike the heterogeneous gene expression profiles found among hESCs, these nhpESCs display remarkably homogeneous profiles (>97%), with full-sibling lines nearly identical (>98.2%). Female nhpESCs express genes distinct from their brother lines; these sensitive analyses are enabled because of the very low background differences. Experimental comparisons among these primate ESCs may prove more reliable than currently available hESCs, since they are akin to inbred mouse strains in which genetic variables are also nearly eliminated. Finally, contrasting the biological similarities among these lines with the heterogeneous hESCs might suggest that additional, more uniform hESC lines are justified. Taken together, pedigreed primate ESCs display homogeneous and reliable expression profiles. These similarities to mouse ESCs suggest that heterogeneities found among hESCs likely result from their disparate origins rather than intrinsic biological limitations with primate embryonic stem cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/physiology , Pedigree , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Female , Macaca mulatta , Male
15.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit 1A.1, 2007 Jun.
Article in English | MEDLINE | ID: mdl-18785157

ABSTRACT

Embryonic stem (ES) cells are a powerful research tool enabling the generation of mice with custom genetics, the study of the earliest stages of mammalian differentiation in vitro and, with the isolation of human ES cells, the potential of cell-based therapies for a number of diseases including Parkinson's and Type 1 diabetes. ES cells isolated from nonhuman primates (nhpES cells) offer the opportunity to ethically test the developmental potential of primate ES cells in chimeric offspring. If these cells have similar potency to mouse ES cells, this may open a new era of primate models of human disease. Nonhuman primates are the perfect model system for the preclinical testing of ES cell-derived therapies. In this unit, we describe methods for the derivation and characterization of nonhuman primate ES cells. With these protocols, the investigator will be able to isolate nhpES cells and perform the necessary tests to confirm the pluripotent phenotype.


Subject(s)
Embryonic Stem Cells/cytology , Primates/embryology , Animals , Base Sequence , Cell Culture Techniques , Cell Separation , DNA Primers/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Gene Expression , Immunohistochemistry , Karyotyping , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Primates/genetics , Primates/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Teratoma/etiology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...