Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
PLoS One ; 10(2): e0116956, 2015.
Article in English | MEDLINE | ID: mdl-25706128

ABSTRACT

Previously, in an attempt to isolate stem cells that would be capable of regenerating injured skeletal muscle, we cultured cells derived from muscle, non-adherently, in serum-free media. As a result of the culture conditions used, these cells formed spheres, and thus were referred to as myospheres. It was found that myosphere-derived cells expressed Sca-1, a marker that is not typically associated with myogenic cells, and as a result has generated some questions as to the origin of these cells. The goal of this study was to clearly determine the origin of myosphere-derived cells, and in particular to answer the question of whether myospheres contain myogenic cells. To determine if myospheres were composed of myogenic cells without altering the structure of myospheres or the culture conditions used to maintain myospheres, I isolated these cells from yellow fluorescent protein (YFP)-Myf5, YFP-MyoD, and ZsGreen-Pax7 lineage-tracing mice and monitored their growth over time. I found that myospheres do contain myogenic cells, but that these cells are gradually lost over time (within 2 months). Additionally, the use of the lineage-tracing mice gave an interesting perspective into the composition of myospheres. I found that myospheres were composed of two distinct cell types, one that is myogenic (α7 integrin+) and contains cells expressing Myf5, MyoD, and Pax7, and a second that is non-myogenic (α7 integrin-) expressing platelet-derived growth factor receptor alpha (PDGFRα) and Sca-1, both of which have been associated with fibro/adipocyte mesenchymal cells.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Mesenchymal Stem Cells/cytology , Muscle Development/physiology , Muscle, Skeletal/cytology , Animals , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mice , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , PAX7 Transcription Factor/metabolism
2.
BMJ Open ; 4(8): e005178, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25142262

ABSTRACT

OBJECTIVE: To evaluate the effects of Quality and Outcomes Framework (QOF) incentivised case finding for depression on diagnosis and treatment in targeted and non-targeted long-term conditions. DESIGN: Interrupted time series analysis. SETTING: General practices in Leeds, UK. PARTICIPANTS: 65 (58%) of 112 general practices shared data on 37,229 patients with diabetes and coronary heart disease targeted by case finding incentives, and 101,008 patients with four other long-term conditions not targeted (hypertension, epilepsy, chronic obstructive pulmonary disease and asthma). INTERVENTION: Incentivised case finding for depression using two standard screening questions. MAIN OUTCOME MEASURES: Clinical codes indicating new depression-related diagnoses and new prescriptions of antidepressants. We extracted routinely recorded data from February 2002 through April 2012. The number of new diagnoses and prescriptions for those on registers was modelled with a binomial regression, which provided the strength of associations between time periods and their rates. RESULTS: New diagnoses of depression increased from 21 to 94/100,000 per month in targeted patients between the periods 2002-2004 and 2007-2011 (OR 2.09; 1.92 to 2.27). The rate increased from 27 to 77/100,000 per month in non-targeted patients (OR 1.53; 1.46 to 1.62). The slopes in prescribing for both groups flattened to zero immediately after QOF was introduced but before incentivised case finding (p<0.01 for both). Antidepressant prescribing in targeted patients returned to the pre-QOF secular upward trend (Wald test for equivalence of slope, z=0.73, p=0.47); the slope was less steep for non-targeted patients (z=-4.14, p<0.01). CONCLUSIONS: Incentivised case finding increased new depression-related diagnoses. The establishment of QOF disrupted rising trends in new prescriptions of antidepressants, which resumed following the introduction of incentivised case finding. Prescribing trends are of concern given that they may include people with mild-to-moderate depression unlikely to respond to such treatment.


Subject(s)
Coronary Disease/economics , Depression/diagnosis , Diabetes Mellitus/economics , General Practice , Physician Incentive Plans/organization & administration , Physicians, Primary Care/economics , Reimbursement, Incentive/organization & administration , Antidepressive Agents/economics , Coronary Disease/epidemiology , Coronary Disease/psychology , Depression/economics , Depression/epidemiology , Depression/etiology , Diabetes Mellitus/epidemiology , Diabetes Mellitus/psychology , Humans , Interrupted Time Series Analysis , Outcome Assessment, Health Care , Physician Incentive Plans/economics , Prescriptions , Quality Improvement , Quality of Health Care , United Kingdom/epidemiology
3.
Biol Open ; 1(9): 839-47, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-23213478

ABSTRACT

A myosphere cell is a unique type of muscle stem cell that is able to maintain its pre-myogenic state in culture over time. These cells are propagated in culture as free-floating, non-adherent spheres. We believe that the 3-dimensional adhesive cell-cell interactions involved in maintaining the sphere-like myosphere structures are also involved in maintaining their longevity in culture. We found that Sca-1, which is highly expressed by myosphere cells, plays a role in the growth and the formation of the myospheres. In comparing adhesion molecules expressed by 3-dimensionally grown myosphere cells to those expressed by 2-dimensionally grown primary myoblasts, we found that there was a distinct difference in the expression of ß3 integrin. Upon further investigation we discovered that there is an adhesive interaction between Sca-1(+) cells and αVß3 integrin. Here we show that Sca-1(+) cells (myosphere cells and NIH3T3 cells) adhere to αVß3 integrin and that Sca-1(-) cells (primary myoblasts) do not adhere. The interaction between Sca-1 and αVß3 integrin was confirmed using antibody blocking, shRNA knockdown of Sca-1 in Sca-1(+) cells, and by expressing Sca-1 cDNA in Sca-1(-) cells, which demonstrated that the level of adhesion of these cells to αVß3 integrin was dependent on the presence of Sca-1. Additionally, we found that the co-expression of Sca-1 and ß3 resulted in significantly greater adhesion of Sca-1(+) cells to αVß3 integrin. In conclusion, our data indicate that Sca-1 is involved in maintaining the 3-dimensional myosphere cell-cell contacts and that Sca-1 is involved in the binding of cells to αVß3 integrin.

4.
Circ Res ; 109(12): 1363-74, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22034491

ABSTRACT

RATIONALE: Recent work in animal models and humans has demonstrated the presence of organ-specific progenitor cells required for the regenerative capacity of the adult heart. In response to tissue injury, progenitor cells differentiate into specialized cells, while their numbers are maintained through mechanisms of self-renewal. The molecular cues that dictate the self-renewal of adult progenitor cells in the heart, however, remain unclear. OBJECTIVE: We investigate the role of canonical Wnt signaling on adult cardiac side population (CSP) cells under physiological and disease conditions. METHODS AND RESULTS: CSP cells isolated from C57BL/6J mice were used to study the effects of canonical Wnt signaling on their proliferative capacity. The proliferative capacity of CSP cells was also tested after injection of recombinant Wnt3a protein (r-Wnt3a) in the left ventricular free wall. Wnt signaling was found to decrease the proliferation of adult CSP cells, both in vitro and in vivo, through suppression of cell cycle progression. Wnt stimulation exerted its antiproliferative effects through a previously unappreciated activation of insulin-like growth factor binding protein 3 (IGFBP3), which requires intact IGF binding site for its action. Moreover, injection of r-Wnt3a after myocardial infarction in mice showed that Wnt signaling limits CSP cell renewal, blocks endogenous cardiac regeneration and impairs cardiac performance, highlighting the importance of progenitor cells in maintaining tissue function after injury. CONCLUSIONS: Our study identifies canonical Wnt signaling and the novel downstream mediator, IGFBP3, as key regulators of adult cardiac progenitor self-renewal in physiological and pathological states.


Subject(s)
Cell Proliferation , Insulin-Like Growth Factor Binding Protein 3/physiology , Myocytes, Cardiac/physiology , Signal Transduction/physiology , Stem Cells/physiology , Wnt Proteins/physiology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation/drug effects , Female , Heart Ventricles/drug effects , Heart Ventricles/pathology , Homeostasis/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Models, Animal , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Recombinant Proteins/pharmacology , Stem Cells/cytology , Wnt3A Protein/pharmacology
5.
Tissue Eng Part A ; 17(5-6): 607-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20883115

ABSTRACT

Mature adult tissues contain stem cells that express many genes normally associated with the early stage of embryonic development, when maintained in appropriate environments. Cells procured from adult tissues representative of the three germ layers (spinal cord, muscle, and lung), each exhibiting the potential to mature into cells representative of all three germ layers. Cells isolated from adult tissues of different germ layer origin were propagated as nonadherent clusters or spheres that were composed of heterogeneous populations of cells. When the clusters or spheres were dissociated, the cells had the ability to reform new, nonadherent spheres for several generations. When implanted in vivo, in association with biodegradable scaffolds, into immunodeficient mice, tissue containing cells characteristic of the three germ layers was generated. These findings suggest the existence of a population of stem cells in adult tissues that is quite different and distinct from embryonic stem cells that demonstrate a greater potency for differentiation across germ lines than previously believed. Such cells could potentially be as useful as embryonic stem cells in tissue engineering and regenerative medicine.


Subject(s)
Adult Stem Cells/cytology , Germ Layers/cytology , Adult Stem Cells/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Cell Aggregation , Cell Differentiation , Embryo, Mammalian/cytology , Gene Expression Profiling , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/metabolism , Phenotype
6.
Nature ; 467(7313): 318-22, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20844535

ABSTRACT

The ß-haemoglobinopathies are the most prevalent inherited disorders worldwide. Gene therapy of ß-thalassaemia is particularly challenging given the requirement for massive haemoglobin production in a lineage-specific manner and the lack of selective advantage for corrected haematopoietic stem cells. Compound ß(E)/ß(0)-thalassaemia is the most common form of severe thalassaemia in southeast Asian countries and their diasporas. The ß(E)-globin allele bears a point mutation that causes alternative splicing. The abnormally spliced form is non-coding, whereas the correctly spliced messenger RNA expresses a mutated ß(E)-globin with partial instability. When this is compounded with a non-functional ß(0) allele, a profound decrease in ß-globin synthesis results, and approximately half of ß(E)/ß(0)-thalassaemia patients are transfusion-dependent. The only available curative therapy is allogeneic haematopoietic stem cell transplantation, although most patients do not have a human-leukocyte-antigen-matched, geno-identical donor, and those who do still risk rejection or graft-versus-host disease. Here we show that, 33 months after lentiviral ß-globin gene transfer, an adult patient with severe ß(E)/ß(0)-thalassaemia dependent on monthly transfusions since early childhood has become transfusion independent for the past 21 months. Blood haemoglobin is maintained between 9 and 10 g dl(-1), of which one-third contains vector-encoded ß-globin. Most of the therapeutic benefit results from a dominant, myeloid-biased cell clone, in which the integrated vector causes transcriptional activation of HMGA2 in erythroid cells with further increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 microRNAs. The clonal dominance that accompanies therapeutic efficacy may be coincidental and stochastic or result from a hitherto benign cell expansion caused by dysregulation of the HMGA2 gene in stem/progenitor cells.


Subject(s)
Blood Transfusion , Genetic Therapy , HMGA2 Protein/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Adolescent , Blood Cells/cytology , Blood Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Child, Preschool , Clone Cells/metabolism , Gene Expression , Genetic Vectors/genetics , HMGA2 Protein/genetics , Homeostasis , Humans , Lentivirus/genetics , Male , MicroRNAs/genetics , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/genetics , Time Factors , Transcriptional Activation , Young Adult , beta-Thalassemia/metabolism
7.
Exp Cell Res ; 316(12): 1966-76, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20381487

ABSTRACT

The effectiveness of cell-based therapy to treat muscle disease has been hampered by difficulties in isolating, maintaining and propagating the stem cells that are needed for treatment. Here we report the isolation of muscle-derived stem cells from both young and old mice and their propagation over extended periods of time in culture as "free-floating" myospheres. Analysis of these sphere-forming cells showed that they express stem cell antigen-1 (Sca-1), beta1 integrin (CD29), Thy-1 (CD90), and CD34, but did not express CD45, CD31, or myogenic markers (Pax7, Myf5, and MyoD). We found that cells derived from myospheres and then grown adherently (MDACs) behaved similar to primary myoblasts, in that these cells expressed myogenic markers and were able to easily form multinucleated myotubes. Unlike the parental myospheres but analogous to primary myoblasts, MDACs expressed Pax7, Myf5, and MyoD, indicating that the parent myosphere cells were a more primitive type of cell. In support of this we demonstrated that myospheres were also able to differentiate into adipogenic and osteogenic cells in culture, as well as being able to contribute to injured muscle in vivo. In summary, we report that primitive adult muscle stem cells can be easily isolated and sustained in culture as myospheres.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Myoblasts/cytology , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred C57BL , Myoblasts/metabolism
8.
Circ Res ; 103(8): 825-35, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18787193

ABSTRACT

Recently, the side population (SP) phenotype has been introduced as a reliable marker to identify subpopulations of cells with stem/progenitor cell properties in various tissues. We and others have identified SP cells from postmitotic tissues, including adult myocardium, in which they have been suggested to contribute to cellular regeneration following injury. SP cells are identified and characterized by a unique efflux of Hoechst 33342 dye. Abcg2 belongs to the ATP-binding cassette (ABC) transporter superfamily and constitutes the molecular basis for the dye efflux, hence the SP phenotype, in hematopoietic stem cells. Although Abcg2 is also expressed in cardiac SP (cSP) cells, its role in regulating the SP phenotype and function of cSP cells is unknown. Herein, we demonstrate that regulation of the SP phenotype in cSP cells occurs in a dynamic, age-dependent fashion, with Abcg2 as the molecular determinant of the cSP phenotype in the neonatal heart and another ABC transporter, Mdr1, as the main contributor to the SP phenotype in the adult heart. Using loss- and gain-of-function experiments, we find that Abcg2 tightly regulates cell fate and function. Adult cSP cells isolated from mice with genetic ablation of Abcg2 exhibit blunted proliferation capacity and augmented cell death. Conversely, overexpression of Abcg2 is sufficient to enhance cell proliferation, although with a limitation of cardiomyogenic differentiation. In summary, for the first time, we reveal a functional role for Abcg2 in modulating the proliferation, differentiation, and survival of adult cSP cells that goes beyond its distinct role in Hoechst dye efflux.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Myocardium/metabolism , Stem Cells/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Age Factors , Aging/metabolism , Animals , Animals, Newborn , Benzimidazoles/metabolism , Cell Death , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Fluorescent Dyes/metabolism , Male , Mice , Mice, Knockout , Myocardium/cytology , Phenotype , Transduction, Genetic , ATP-Binding Cassette Sub-Family B Member 4
9.
J Hepatol ; 47(1): 74-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17434229

ABSTRACT

BACKGROUND/AIMS: Hepatocyte transplantation and bioartificial liver treatment are attractive alternatives to liver transplantation. The availability of well-characterized human hepatocyte lines facilitates such cell therapies. METHODS: Human hepatocytes were immortalized with a retroviral vector SSR#197 expressing catalytic subunit of human telomerase reverse transcriptase (hTERT) and enhanced green fluorescent protein (EGFP) cDNAs flanked by a pair of loxP recombination targets. Then, Tamoxifen-dependent Cre recombinase was expressed in SSR#197-immortalized hepatocytes. Cre/LoxP recombination was performed in the established cells by simple exposure to 500 nM Tamoxifen for a week. Then, the reverted population of the cells was recovered by EGFP-negative cell sorting and characterized in vitro and in vivo using a pig model of acute liver failure (ALF) induced by d-galactosamine (0.5 g/kg) injection. RESULTS: A human hepatocyte cell line 16T-3 was established. Reverted 16-T3 cells showed the increased expression of hepatic markers in association with enhanced levels of transcriptional factors. Compared to normal human hepatocytes, albumin production and lidocaine-metabolizing activities of reverted 16-T3 cells were 0.32 and 0.50-fold, respectively. Transplantation of reverted 16T-3 cells significantly prolonged the survival of ALF pigs. CONCLUSIONS: Here we demonstrate the usefulness of Cre/LoxP -mediated reversible immortalization of human hepatocytes with Tamoxifen-mediated self-recombination.


Subject(s)
Cell Line, Transformed/transplantation , Hepatocytes/transplantation , Liver Failure/surgery , Animals , Biomarkers/analysis , Cell Line, Transformed/chemistry , Cell Line, Transformed/cytology , Hepatocytes/chemistry , Hepatocytes/drug effects , Humans , Integrases/genetics , Liver Failure/pathology , Recombination, Genetic , Retroviridae/genetics , Sus scrofa , Tamoxifen/pharmacology , Treatment Outcome
10.
Retrovirology ; 4: 96, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-18163907

ABSTRACT

BACKGROUND: The structural and enzymatic proteins of the human immunodeficiency virus (HIV) are initially generated as two long polyproteins encoded from overlapping reading frames, one producing the structural proteins (Gag) and the second producing both structural and enzymatic proteins (Gag-Pol). The Gag to Gag-Pol ratio is critical for the proper assembly and maturation of viral particles. To minimize the risk of producing a replication competent lentivirus (RCL), we developed a "super-split" lentiviral packaging system in which Gag was separated from Pol with minimal loss of transducibility by supplying protease (PR) in trans independently of both Gag and Pol. RESULTS: In developing this "super-split" packaging system, we incorporated several new safety features that include removing the Gag/Gag-Pol frameshift, splitting the Gag, PR, and reverse transcriptase/integrase (RT/IN) functions onto separate plasmids, and greatly reducing the nucleotide sequence overlap between vector and Gag and between Gag and Pol. As part of the construction of this novel system, we used a truncated form of the accessory protein Vpr, which binds the P6 region of Gag, as a vehicle to deliver both PR and RT/IN as fusion proteins to the site of viral assembly and budding. We also replaced wt PR with a slightly less active T26S PR mutant in an effort to prevent premature processing and cytoxicity associated with wt PR. This novel "super-split" packaging system yielded lentiviral titers comparable to those generated by conventional lentiviral packaging where Gag-Pol is supplied intact (1.0 x 106 TU/ml, unconcentrated). CONCLUSION: Here, we were able to create a true "split-function" lentiviral packaging system that has the potential to be used for gene therapy applications. This novel system incorporates many new safety features while maintaining high titers. In addition, because PR is supplied in trans, this unique system may also provide opportunities to examine viral protein processing and maturation.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Virus Assembly , Cell Line , Humans , Lentivirus/growth & development , Lentivirus/physiology , Plasmids , Viral Proteins/genetics , Viral Proteins/metabolism
11.
FASEB J ; 20(13): 2242-50, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17077301

ABSTRACT

Adenosine has been widely associated with hypoxia of many origins, including those associated with inflammation and tumorogenesis. A number of recent studies have implicated metabolic control of adenosine generation at sites of tissue hypoxia. Here, we examine adenosine receptor control and amplification of signaling through transcriptional regulation of endothelial and epithelial adenosine receptors. Initial studies confirmed previous findings indicating selective induction of human adenosine A2B receptor (A2BR) by hypoxia. Analysis of the cloned human A2BR promoter identified a functional hypoxia-responsive region, including a functional binding site for hypoxia-inducible factor (HIF) within the A2BR promoter. Further studies examining HIF-1alpha DNA binding and HIF-1alpha gain and loss of function confirmed strong dependence of A2BR induction by HIF-1alpha in vitro and in vivo mouse models. Additional studies in endothelia overexpressing full-length A2BR revealed functional phenotypes of increased barrier function and enhanced angiogenesis. Taken together, these results demonstrate transcriptional coordination of A2BR by HIF-1alpha and amplified adenosine signaling during hypoxia. These findings may provide an important link between hypoxia and metabolic conditions associated with inflammation and angiogenesis.


Subject(s)
Cell Hypoxia/physiology , Endothelium, Vascular/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Intestinal Mucosa/physiology , Receptor, Adenosine A2B/genetics , Base Sequence , Cell Line , Cells, Cultured , Chromatin/genetics , Cloning, Molecular , DNA Primers , Endothelium, Vascular/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestinal Mucosa/cytology , Molecular Sequence Data , Neovascularization, Physiologic , Oligonucleotide Array Sequence Analysis , Phenotype , Promoter Regions, Genetic , RNA, Small Interfering/genetics
12.
Int J Oncol ; 28(4): 815-21, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16525629

ABSTRACT

Endothelial progenitor cells (EPCs) contribute to blood vessel formation in ischemic and tumorous tissues, but comprise only a small population in circulation. We attempted to immortalize putative EPCs from human cord blood. Human CD34+ cord blood cells were cultured in the presence of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF), and transfected with a retroviral vector encoding the simian virus 40 large T (SV40T) antigen. This resulted in the immortalization of cord blood cells, leading to the establishment of several cell lines. One of these lines, HYCEC-1, exhibited a phenotype characteristic of the endothelial lineage, including expression of von Willebrand factor and VEGF receptor-2 (VEGFR-2/KDR/Flk-1) and uptake of acetylated-low density lipoprotein. Flow cytometric analysis revealed that HYCEC-1 cells were strongly positive for CD31 and CD146, moderately positive for CD144, weakly positive for CD133 and CD34, and negative for CD14 and CD45. HYCEC-1 cells formed capillary-like structures on basement matrix gel in vitro. Upon transplantation into the ischemic hind limb of nude rats, HYCEC-1 cells efficiently participated in neovascularization and augmented blood flow. The immortalized HYCEC-1 cells are suggested to be a class of EPCs that can efficiently participate in postnatal neovasculogenesis in the ischemic hind limb, and may also be a useful tool for studying tumor vessel formation.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Endothelial Cells/metabolism , Neovascularization, Pathologic , Stem Cells/metabolism , AC133 Antigen , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Blood Flow Velocity , CD146 Antigen/analysis , Cadherins/analysis , Cell Line , Cell Line, Transformed , Cell Transformation, Viral/genetics , Cell Transformation, Viral/immunology , Cell Transplantation/methods , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/immunology , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/metabolism , Fibroblast Growth Factor 2/pharmacology , Glycoproteins/analysis , Hindlimb/blood supply , Hindlimb/surgery , Humans , Ischemia/physiopathology , Neovascularization, Physiologic , Peptides/analysis , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Rats , Rats, Inbred F344 , Rats, Nude , Stem Cells/drug effects , Stem Cells/immunology , Transfection , Vascular Endothelial Growth Factor A/pharmacology
13.
J Exp Med ; 202(11): 1493-505, 2005 Dec 05.
Article in English | MEDLINE | ID: mdl-16330813

ABSTRACT

Extracellular adenosine (Ado) has been implicated as central signaling molecule during conditions of limited oxygen availability (hypoxia), regulating physiologic outcomes as diverse as vascular leak, leukocyte activation, and accumulation. Presently, the molecular mechanisms that elevate extracellular Ado during hypoxia are unclear. In the present study, we pursued the hypothesis that diminished uptake of Ado effectively enhances extracellular Ado signaling. Initial studies indicated that the half-life of Ado was increased by as much as fivefold after exposure of endothelia to hypoxia. Examination of expressional levels of the equilibrative nucleoside transporter (ENT)1 and ENT2 revealed a transcriptionally dependent decrease in mRNA, protein, and function in endothelia and epithelia. Examination of the ENT1 promoter identified a hypoxia inducible factor 1 (HIF-1)-dependent repression of ENT1 during hypoxia. Using in vitro and in vivo models of Ado signaling, we revealed that decreased Ado uptake promotes vascular barrier and dampens neutrophil tissue accumulation during hypoxia. Moreover, epithelial Hif1alpha mutant animals displayed increased epithelial ENT1 expression. Together, these results identify transcriptional repression of ENT as an innate mechanism to elevate extracellular Ado during hypoxia.


Subject(s)
Adenosine/pharmacology , Down-Regulation/drug effects , Equilibrative Nucleoside Transporter 1/biosynthesis , Equilibrative-Nucleoside Transporter 2/biosynthesis , Signal Transduction/drug effects , Vasodilator Agents/pharmacology , Adenosine/metabolism , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line , Down-Regulation/physiology , Epithelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1 , Neutrophils/metabolism , Signal Transduction/physiology , Vasodilator Agents/metabolism
14.
Nat Biotechnol ; 23(10): 1274-82, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186810

ABSTRACT

A human pancreatic beta-cell line that is functionally equivalent to primary beta-cells has not been available. We established a reversibly immortalized human beta-cell clone (NAKT-15) by transfection of primary human beta-cells with a retroviral vector containing simian virus 40 large T-antigen (SV40T) and human telomerase reverse transcriptase (hTERT) cDNAs flanked by paired loxP recombination targets, which allow deletion of SV40T and TERT by Cre recombinase. Reverted NAKT-15 cells expressed beta-cell transcription factors (Isl-1, Pax 6, Nkx 6.1, Pdx-1), prohormone convertases 1/3 and 2, and secretory granule proteins, and secreted insulin in response to glucose, similar to normal human islets. Transplantation of NAKT-15 cells into streptozotocin-induced diabetic severe combined immunodeficiency mice resulted in perfect control of blood glucose within 2 weeks; mice remained normoglycemic for longer than 30 weeks. The establishment of this cell line is one step toward a potential cure of diabetes by transplantation.


Subject(s)
Cell Culture Techniques/methods , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/physiology , Tissue Engineering/methods , Animals , Cell Line , Cell Proliferation , Genetic Enhancement/methods , Humans , Mice , Mice, SCID , Treatment Outcome
15.
J Clin Invest ; 114(7): 953-62, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15467834

ABSTRACT

Transplantation of genetically corrected autologous hematopoietic stem cells is an attractive approach for the cure of sickle-cell disease and beta-thalassemia. Here, we infected human cord blood cells with a self-inactivating lentiviral vector encoding an anti-sickling betaA-T87Q-globin transgene and analyzed the transduced progeny produced over a 6-month period after transplantation of the infected cells directly into sublethally irradiated NOD/LtSz-scid/scid mice. Approximately half of the human erythroid and myeloid progenitors regenerated in the mice containing the transgene, and erythroid cells derived in vitro from these in vivo-regenerated cells produced high levels of betaA-T87Q-globin protein. Linker-mediated PCR analysis identified multiple transgene-positive clones in all mice analyzed with 2.1 +/- 0.1 integrated proviral copies per cell. Genomic sequencing of vector-containing fragments showed that 86% of the proviral inserts had occurred within genes, including several genes implicated in human leukemia. These findings indicate effective transduction of very primitive human cord blood cells with a candidate therapeutic lentiviral vector resulting in the long-term and robust, erythroid-specific production of therapeutically relevant levels of beta-globin protein. However, the frequency of proviral integration within genes that regulate hematopoiesis points to a need for additional safety modifications.


Subject(s)
Fetal Blood/cytology , Genetic Vectors/metabolism , Globins/metabolism , Hematopoietic Stem Cells/physiology , Lentivirus/metabolism , Transduction, Genetic , Animals , Cell Lineage , Cell Transplantation , Cells, Cultured , Chromosomes, Human , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Globins/genetics , Hematopoietic Stem Cells/cytology , Humans , Lentivirus/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Transgenes
16.
Transplantation ; 77(9): 1357-65, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15167590

ABSTRACT

BACKGROUND AND AIMS: Liver endothelial cells (LECs) perform an essential role in important pathophysiologic functions in the liver. Establishment of a human LEC line facilitates advances in LEC research. Here, we present immortalization of human LECs using retroviral gene transfer of simian virus 40 large T antigen (SV40T) and human telomerase reverse transcriptase (hTERT). We also demonstrate excision of SV40T and hTERT with TAT-mediated Cre/loxP recombination and subsequent cell sorting. METHODS: First, human LECs were transduced with a retroviral vector somatostatin receptor (SSR)#69 expressing SV40T and hygromycin-resistance genes flanked by a pair of loxA recombination targets. Then, cells were retrovirally superinfected with SSR#197 encoding hTERT and green fluorescent protein (GFP) cDNAs that were intervened by two loxBs. One SV40T-and hTERT-immortalized LEC clone, TMNK-1, was established and analyzed for its biologic characteristics. RESULTS: The cells were hygromycin-resistant and uniformly positive for GFP expression. TMNK-1 expressed EC markers, including factor VIII, vascular endothelial growth factor receptors (flt-1, KDR/Flk-1), and CD34, showed uptake of Di-I-acetylated-low-density lipoprotein and angiogenic potential in Matrigel assays. After lipopolysaccharide treatment, TMNK-1 produced tumor necrosis factor (TNF)-alpha and interleukin (IL)-6 and exhibited increased expression of intra-cellular adhesive molecule-1, vascular cellular adhesive molecule-1, and VE-cadherin. After treatment with TAT-Cre recombinase fusion protein, approximately 60% of TMNK-1 was negative for GFP expression, and subsequent cell sorting of this population for GFP allowed for collection of the reverted form of TMNK-1. CONCLUSIONS: This study demonstrates the utility and efficiency of the reversible immortalization procedure to expand primary human LECs for basic studies.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Cell Line, Transformed/cytology , Endothelial Cells/cytology , Liver/cytology , Telomerase/genetics , Animals , Antigens, CD , Biomarkers , Cadherins/genetics , Cell Line, Transformed/physiology , Cell Separation , DNA-Binding Proteins , E-Selectin/genetics , Gene Products, tat/genetics , Humans , Integrases/genetics , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Physiologic , Receptors, Cell Surface/genetics , Retroviridae/genetics , Toll-Like Receptors , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Viral Proteins/genetics
17.
Transplantation ; 77(3): 446-51, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14966424

ABSTRACT

BACKGROUND: Cholangiocytes perform an essential role in important pathophysiologic functions in the liver. Establishment of a human cholangiocyte line facilitates advances in cholangiocyte research and clinical applications for cell therapies. Here, we describe the immortalization of human cholangiocytes using serial transfection of simian virus 40 large T (SV40T) followed by human telomerase reverse transcriptase (hTERT). METHODS: SV40T-transduced human liver OUMS-21 cells were superinfected with a retroviral vector SSR#197 encoding hTERT and green fluorescent protein (GFP) cDNAs. Resulting cell lines were evaluated for gene expression, functional cholangiogenic characteristics in vitro and in vivo, and response to lipopolysaccharide (LPS). RESULTS: One of the SV40T- and hTERT-immortalized cholangiocyte clones, MMNK-1, was established. MMNK-1 expressed cholangiocyte markers, including cytokeratin (CK)-7 and -19 and exhibited cholangiogenic tubule formation in a Matrigel assay. When transplanted into the immunodeficient mice, MMNK-1 cells developed bile duct-like structures in the spleen. After LPS treatment, MMNK-1 cells produced interleukin-6 and failed to form well-developed tubular structures in Matrigel. CONCLUSION: We have established an immortalized cholangiocyte cell line, MMNK-1, using SV40T and hTERT transduction.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Cell Line, Transformed , Liver/cytology , Telomerase/genetics , Transfection , Animals , Biocompatible Materials , Biomarkers/analysis , Cell Differentiation , Cell Transplantation , Collagen , DNA-Binding Proteins , Drug Combinations , Humans , Interleukin-6/biosynthesis , Laminin , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, SCID , Microscopy, Electron, Scanning , Proteoglycans , Telomerase/metabolism
18.
Diabetes ; 53(1): 105-12, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14693704

ABSTRACT

Type 1 diabetes results from the destruction of insulin-producing pancreatic beta-cells by a beta-cell-specific autoimmune process. Although converting other cell types into insulin-producing cells may compensate for the loss of the beta-cell mass while evading beta-cell-specific T-cell responses, proof-of-principle of this approach in large animal models is lacking. This investigation was initiated to determine whether an insulin-producing human hepatocyte line can control diabetes when transplanted into totally pancreatectomized diabetic pigs. We established a reversibly immortalized human hepatocyte line, YOCK-13, by transferring a human telomerase reverse transcriptase cDNA and a drug-inducible Cre recombinase cassette, followed by cDNA for a modified insulin under the control of the L-type pyruvate kinase (L-PK) promoter. YOCK-13 cells produced small amounts of modified insulin and no detectable endogenous L-PK at low glucose concentrations, whereas they produced large amounts of both modified insulin and L-PK in response to high glucose concentrations. Xenotransplantation of YOCK-13 cells via the portal vein into immunosuppressed, totally pancreatectomized pigs decreased hyperglycemia and prolonged survival without adverse effects such as portal thrombosis, liver necrosis, pulmonary embolism, and tumor development. We suggest that this reversibly immortalized, insulin-secreting human hepatocyte line may overcome the shortage of donor pancreata for islet transplantation into patients with type 1 diabetes.


Subject(s)
Hepatocytes/transplantation , Hyperglycemia/prevention & control , Insulin/metabolism , Animals , Cell Line , DNA-Binding Proteins , Disease Models, Animal , Hepatocytes/metabolism , Humans , Insulin Secretion , Pancreatectomy , Portal Vein , Promoter Regions, Genetic , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Survival Analysis , Swine , Telomerase/genetics , Telomerase/metabolism , Transplantation, Heterologous/physiology
19.
Int J Oncol ; 23(4): 925-32, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12963970

ABSTRACT

Human marrow-derived mesenchymal stem cells (MSC), which have the potential to differentiate into mesenchymal tissues, such as bone, cartilage, adipose and bone marrow stroma, were transduced with a retroviral vector carrying the simian virus 40 large T antigen, hygromycin-resistant gene and herpes simplex virus thymidine kinase gene, that can be excised by Cre/loxP site-specific recombination. This resulted in establishment of an MSC cell line, HMSC-1, which retained original surface characteristics and differentiation potential, and exhibited a higher proliferative capacity than parental cells. HMSC-1 expressed mRNAs of BMP-4, Jagged-1, and SCF that are known to promote hematopoiesis. Human CB CD34+ hematopoietic progenitor cells (HPC) cultured on a layer of HMSC-1 cells showed high expansion of CD34+CD38- immature HPC, capable of reconstituting human hematopoiesis in non-obese diabetic/severe combined immunodeficient disease (NOD/SCID) mice. This cell line may be of value for developing strategies for ex vivo expansion of human HPC.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Bone Marrow Cells/cytology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hygromycin B/analogs & derivatives , Mesoderm/cytology , Stem Cells/cytology , ADP-ribosyl Cyclase/biosynthesis , ADP-ribosyl Cyclase 1 , Animals , Antigens, CD/biosynthesis , Antigens, CD34/biosynthesis , Blotting, Western , Cell Culture Techniques/methods , Cell Differentiation , Cinnamates/pharmacology , Coculture Techniques , Colony-Forming Units Assay , Flow Cytometry , Humans , Hygromycin B/pharmacology , Membrane Glycoproteins , Mice , Mice, SCID , Models, Genetic , Phenotype , RNA, Messenger/metabolism , Recombination, Genetic , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
20.
Transplantation ; 75(11): 1873-80, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12811248

ABSTRACT

BACKGROUND: Maintenance of liver-specific functions has been shown to be stabilized by co-cultivation of hepatocytes with hepatic stellate cells (HSC). Because the limited lifespan of human HSC is a major hurdle to their use, the authors report here the amplification of human HSC populations in vitro by retroviral transfer of human telomerase reverse transcriptase (hTERT). METHODS: Human HSC strain LI 90 cells were transduced with a retroviral vector SSR#197 expressing hTERT and green fluorescent protein (GFP) cDNA flanked by a pair of loxP. TWNT-1, one of SSR#197-immortalized HSC, was characterized. Differentiated liver functions were evaluated in an immortalized human hepatocyte NKNT-3-TWNT-1 co-culture system. RESULTS: TWNT-1 cells showed differential functions of HSC, including uptake of acetylated low-density lipoproteins and synthesis of collagen type I and hepatocyte growth factor. Efficient excision of the retrovirally transferred hTERT and GFP cDNAs was achieved by TAT-mediated expression of the Cre recombinase and subsequent GFP-negative cell sorting. When co-cultured with TWNT-1 cells, NKNT-3 increased protein expression of the detoxifying cytochrome P450-associated protein isoenzymes 3A4 and 2C9 and urea synthesis. CONCLUSIONS: TWNT-1 cells could be valuable in the study of integrated liver functions and contribute to the optimization of liver cell therapies and bioartificial livers.


Subject(s)
Hepatocytes/cytology , Liver, Artificial , Acetylation , Actins/genetics , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Carbocyanines , Cell Division , Cell Line, Transformed , Cholesterol, LDL/pharmacokinetics , Coculture Techniques , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins , Endocytosis , Female , Fluorescent Dyes , Gene Expression , Gene Products, tat/genetics , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Integrases/genetics , Luminescent Proteins/genetics , Mice , Mice, SCID , Middle Aged , Receptor, Platelet-Derived Growth Factor beta/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Urea/metabolism , Viral Proteins/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...