Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Int J Part Ther ; 7(4): 19-28, 2021.
Article in English | MEDLINE | ID: mdl-33829070

ABSTRACT

PURPOSE: To investigate dosimetric implications of biodegradable Biozorb (BZ) markers for proton accelerated partial breast irradiation (APBI) plans. MATERIALS AND METHODS: Six different BZs were placed within in-house breast phantoms to acquire computed tomography (CT) images. A contour correction method with proper mass density overriding for BZ titanium clip and surrounding tissue was applied to minimize inaccuracies found in the CT images in the RayStation planning system. Each breast phantom was irradiated by a monoenergetic proton beam (103.23 MeV and 8×8 cm2) using a pencil-beam scanning proton therapy system. For a range perturbation study, doses were measured at 5 depths below the breast phantoms by using an ionization chamber and compared to the RayStation calculations with 3 scenarios for the clip density: the density correction method (S1: 1.6 g/cm3), raw CT (S2), and titanium density (S3: 4.54 g/cm3). For the local dose perturbation study, the radiographic EDR2 film was placed at 0 and 2 cm below the phantoms and compared to the RayStation calculations. Clinical effects of the perturbations were retrospectively examined with 10 APBI plans for the 3 scenarios (approved by our institutional review board). RESULTS: In the range perturbation study, the S1 simulation showed a good agreement with the chamber measurements, while excess pullbacks of 1∼2 mm were found in the S2 and S3 simulations. The film study showed local dose shadowing and perturbation by the clips that RayStation could not predict. In the plan study, no significant differences in the plan quality were found among the 3 scenarios. However, substantial range pullbacks were observed for S3. CONCLUSION: The density correction method could minimize the dose and range difference between measurement and RayStation prediction. It should be avoided to simply override the known physical density of the BZ clips for treatment planning owing to overestimation of the range pullback.

2.
Br J Cancer ; 106(3): 553-61, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22223089

ABSTRACT

BACKGROUND: The incidence of malignant melanoma is increasing faster than that for any other cancer. Histological examination of skin excision biopsies remains the standard method for melanoma diagnosis and prognosis. Significant morphological overlap between benign and malignant lesions complicates diagnosis, and tumour thickness is not always an accurate predictor of prognosis. METHODS: To identify improved molecular markers to support histological examination, we used microarray analysis of formalin-fixed and paraffin-embedded samples from different stages of melanomagenesis to identify differentially expressed microRNAs (miRNAs). Differential expression was validated by qRT-PCR, and functional studies were carried out after transfection of miRNA precursors or inhibitors into melanoma cells to modulate miRNA expression. RESULTS: In all, 20 miRNAs showed highly significant differential expression between benign naevi and either primary or metastatic melanomas, the majority being downregulated in melanoma, whereas only 2 miRNAs, namely miR-203 and miR-205, were differentially expressed between primary and metastatic melanomas. In functional in vitro assays, overexpression of miR-200c and miR-205 inhibited anchorage-independent colony formation and overexpression of miR-211 inhibited both anchorage-independent colony formation and invasion. CONCLUSION: We have identified a series of differentially expressed miRNAs that could be useful as diagnostic or prognostic markers for melanoma and have shown that three miRNAs (namely miR-200c, miR-205 and miR-211) act as tumour suppressors.


Subject(s)
Melanoma/genetics , MicroRNAs/genetics , Skin Neoplasms/genetics , Biomarkers , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Melanoma/metabolism , MicroRNAs/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/metabolism
3.
Oncogene ; 31(19): 2412-22, 2012 May 10.
Article in English | MEDLINE | ID: mdl-21996734

ABSTRACT

The incidence of malignant melanoma is growing rapidly worldwide and there is still no effective therapy for metastatic disease. Melanoma is the second most common cancer among young adults in the UK, where incidence rates have more than quadrupled since the 1970s. Increased expression of a number of DNA repair genes has been reported in melanoma and this likely contributes to its extreme resistance to conventional DNA-damaging chemotherapeutics. One such chemotherapeutic that is effective against a range of other cancers, but not melanoma, is cisplatin. The DNA repair proteins ERCC1 and XPF are needed to remove cisplatin-induced DNA damage and we have investigated the response of these proteins to cisplatin in melanoma. The expression of both genes is induced by cisplatin. Use of a MEK inhibitor showed that ERCC1, but not XPF induction was regulated by the mitogen-activated protein kinase (MAPK) pathway, with reduction in expression of DUSP6, the phosphatase that inactivates the extracellular signal-regulated kinase (ERK), being particularly important. DUSP6 overexpression prevented cisplatin induction of both ERCC1 and XPF, resulting in increased sensitivity to cisplatin. A novel ERCC1 mRNA was found that initiated upstream of the normal transcription initiation site, and was strongly regulated by both cisplatin and the MAPK pathway and its role in cisplatin resistance merits further study. The cisplatin induction of ERCC1 and XPF provides important insights into the resistance of melanoma to DNA-damaging chemotherapeutics, which is one of the major obstacles to melanoma treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA Repair/drug effects , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/drug effects , Endonucleases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/genetics , Skin Neoplasms/genetics , Cell Line, Tumor , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 6/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , Signal Transduction/drug effects
4.
Philos Trans R Soc Lond B Biol Sci ; 366(1575): 2307-11, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21727136

ABSTRACT

Stem cells with the potential to form many different cell types are actively studied for their possible use in cell replacement therapies for several diseases. In addition, the differentiated derivatives of stem cells are being used as reagents to test for drugs that slow or correct disease phenotypes found in several degenerative diseases. This paper explores these approaches in the context of type 1 or juvenile diabetes, pointing to recent successes as well as the technical and theoretical challenges that lie ahead in the path to new treatments and cures.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Stem Cell Transplantation/methods , Humans , Insulin-Secreting Cells/physiology
5.
Transgenic Res ; 20(1): 109-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20407824

ABSTRACT

The nucleotide excision repair pathway deals with UV-induced DNA damage. The tissue that receives by far the greatest exposure to UV is the skin and we have investigated the possibility that expression of the nucleotide excision repair gene, Ercc1, may display different properties in the skin to deal with a more demanding role in that tissue. ERCC1, in a complex with XPF, is the structure--specific endonuclease responsible for incising 5' to the UV-induced lesion. We identified a novel Ercc1 mRNA in mouse skin that originates from an alternative upstream promoter. Levels of this skin-specific transcript were low in embryonic skin and increased rapidly after birth, but there was no induction by UV, either in adult skin, or in a cultured keratinocyte model. Levels of the skin-specific Ercc1 transcript were higher in albino than pigmented mouse strains, but there was no difference in ERCC1 protein levels and the expression of the skin-specific transcript was found to be determined by the Ercc1 gene sequence rather than by coat pigmentation. Using an Ercc1 transgene the promoter for the skin-specific transcript was mapped to a region around 400 bp upstream of the normal promoter, where a transposable element with known promoter activity was found in albino but not in pigmented strains.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Promoter Regions, Genetic , Skin/metabolism , Transcription, Genetic , 5' Untranslated Regions , Animals , Base Sequence , Cell Line , Cells, Cultured , DNA-Binding Proteins/chemistry , Endonucleases/chemistry , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Skin/cytology , Transgenes , Ultraviolet Rays
6.
Science ; 322(5909): 1811-5, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19095934

ABSTRACT

Nuclear reprogramming describes a switch in gene expression of one kind of cell to that of another unrelated cell type. Early studies in frog cloning provided some of the first experimental evidence for reprogramming. Subsequent procedures included mammalian somatic cell nuclear transfer, cell fusion, induction of pluripotency by ectopic gene expression, and direct reprogramming. Through these methods it becomes possible to derive one kind of specialized cell (such as a brain cell) from another, more accessible, tissue (such as skin) in the same individual. This has potential applications for cell replacement without the immunosuppression treatments that are required when cells are transferred between genetically different individuals. This article provides some background to this field, a discussion of mechanisms and efficiency, and comments on prospects for future nuclear reprogramming research.


Subject(s)
Cellular Reprogramming , Animals , Cell Dedifferentiation , Cell Differentiation , Cell Fusion , Cell Lineage , Cloning, Organism , DNA/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Gene Expression , Humans , Male , Nuclear Transfer Techniques , Oocytes/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Article in English | MEDLINE | ID: mdl-19478324

ABSTRACT

Diabetes is a leading health problem of the world and its prevalence continues to rise. With Type I diabetes, and in some patients with Type II, the lack of insulin can be counterbalanced by providing new beta (insulin-producing) cells. For Type I diabetes, treating the autoimmune attack remains a serious challenge. Several strategies to produce new beta cells have been proposed. These include differentiation from embryonic stem cells, proliferation of existing adult beta cells, derivation from putative adult progenitors/stem cells, and reprogramming of non-beta cells to beta cells. Each of these strategies has distinct merits and risks, and they are at different stages of understanding and development. In particular, the approach based on differentiation from embryonic stem cells has had strong support and in recent years has made notable progress. Nevertheless, significant hurdles remain to transform the current research into future therapies. To expedite this transformation, we believe particular emphasis should be placed on overcoming key knowledge gaps in beta-cell biology, developing strategies that produce patient-specific beta cells, and carefully addressing potential treatment-related complications or limitations.


Subject(s)
Cell Differentiation/physiology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Cell Differentiation/genetics , Cell Line , Cell Proliferation , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Diabetes Mellitus/therapy , Embryonic Development/genetics , Embryonic Development/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Humans , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Islets of Langerhans/physiology , Signal Transduction
9.
Oncogene ; 25(47): 6229-38, 2006 Oct 12.
Article in English | MEDLINE | ID: mdl-16682947

ABSTRACT

Ercc1 has an essential role in the nucleotide excision repair (NER) pathway that protects against ultraviolet (UV)-induced DNA damage and is also involved in additional repair pathways. The premature death of simple Ercc1 mouse knockouts meant that we were unable to study the role of Ercc1 in the skin. To do this, we have used the Cre-lox system to generate a skin-specific Ercc1 knockout. With a Cre transgene under control of the bovine keratin 5 promoter we achieved 100% recombination of the Ercc1 gene in the epidermis. Hairless mice with Ercc1-deficient skin were hypersensitive to the short-term effects of UV irradiation, showing a very low minimal erythemal dose and a dramatic hyperproliferative response. Ultraviolet-irradiated mice with Ercc1-deficient skin developed epidermal skin tumours much more rapidly than controls. These tumours appeared to arise earlier in actinic progression and grew more rapidly than tumours on control mice. These responses are more pronounced than have been reported for other NER-deficient mice, demonstrating that Ercc1 has a key role in protecting against UV-induced skin cancer.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/physiology , Endonucleases/physiology , Epidermis/enzymology , Neoplasms, Radiation-Induced/enzymology , Skin Neoplasms/enzymology , Ultraviolet Rays/adverse effects , Animals , DNA/radiation effects , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Progression , Endonucleases/deficiency , Endonucleases/genetics , Epidermis/pathology , Epidermis/radiation effects , Female , Gene Targeting , Genes, Lethal , Integrases , Male , Mice , Mice, Hairless , Mice, Knockout , Neoplasms, Radiation-Induced/genetics , Organ Specificity , Skin Neoplasms/genetics , Transgenes
11.
Br J Cancer ; 89(2): 333-7, 2003 Jul 21.
Article in English | MEDLINE | ID: mdl-12865926

ABSTRACT

DNA repair has an essential role in protecting the genome from damage by endogenous and environmental agents. Polymorphisms in DNA repair genes and differences in repair capacity between individuals have been widely documented. For colorectal cancer, the loss of mismatch repair gene activity is a key genetic determinant. Nucleotide excision repair (NER), recombination repair (RR) and base excision repair (BER) pathways have critical roles in protection against other cancers, and we wished to investigate their role in colorectal cancer. We have compared the frequency of polymorphisms in the NER genes, XPD, XPF, XPG, ERCC1; in the BER gene, XRCC1; and in the RR gene, XRCC3; in colorectal cancer patients and in a control group. No significant associations were found for any of the NER gene polymorphisms or for the XRCC1 polymorphism. The C allele (position 18067) of the XRCC3 gene was weakly but significantly associated with colorectal cancer (odds ratio 1.52, 95% confidence interval 1.04-2.22, P=0.03). For all patients who were heterozygous for any of the repair genes studied, tumour tissue was investigated for loss of heterozygosity (LOH). Only one example of LOH was found for all the genes examined. From the association and LOH data, we conclude that these genes do not have an important role in protection against colorectal carcinogenesis.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Repair/genetics , DNA-Binding Proteins/genetics , Polymorphism, Genetic , Cell Transformation, Neoplastic , Genotype , Humans , Loss of Heterozygosity , Odds Ratio , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , X-ray Repair Cross Complementing Protein 1
12.
Nucleic Acids Res ; 29(22): 4541-50, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11713303

ABSTRACT

The ERCC1 gene is essential for the repair of UV-induced DNA damage. Unlike most genes in the nucleotide excision repair (NER) pathway, ERCC1 is also involved in recombinational repair. Perhaps for this reason, ERCC1 knockout mice are not a model for the human NER deficiency disorder, xeroderma pigmentosum. Instead, ERCC1 null mice are severely runted and die before weaning from liver failure with accelerated hepatocyte polyploidy that is more reminiscent of a premature ageing disorder. To permit study of the role of ERCC1 in other tissues we have corrected the liver ERCC1 deficiency with a transgene under the control of a liver-specific promoter. The transgene alleviated runting and extended the lifespan. The elevated level of oxidative DNA damage and premature liver polyploidy were reversed and liver function was corrected. A widespread mitochondrial dysfunction was identified and an essential role for ERCC1 in the kidney was also revealed with transgene-containing ERCC1-deficient animals going on to die of renal failure. The nuclei of kidney proximal tubule cells became polyploid in a similar way to the premature liver polyploidy observed in younger ERCC1-deficient animals. We believe that this is a response to the accumulation of endogenous DNA damage in these particularly susceptible tissues which cannot be repaired in ERCC1-deficient animals.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins , Endonucleases , Guanosine/analogs & derivatives , Liver/metabolism , Proteins/genetics , Transgenes/genetics , Animals , Blotting, Northern , Cell Nucleus/metabolism , DNA/metabolism , DNA Damage , Female , Gene Expression , Genotype , Guanosine/metabolism , Kidney/pathology , Kidney/physiopathology , Lactic Acid/blood , Liver/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/physiology , Oxidative Stress , Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Survival Rate , Time Factors
13.
Science ; 294(5542): 564-7, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11577200

ABSTRACT

Blood vessels supply developing organs with metabolic sustenance. Here, we demonstrate a role for blood vessels as a source of developmental signals during pancreatic organogenesis. In vitro experiments with embryonic mouse tissues demonstrate that blood vessel endothelium induces insulin expression in isolated endoderm. Removal of the dorsal aorta in Xenopus laevis embryos results in the failure of insulin expression in vivo. Furthermore, using transgenic mice, we show that ectopic vascularization in the posterior foregut leads to ectopic insulin expression and islet hyperplasia. These results indicate that vessels not only provide metabolic sustenance, but also provide inductive signals for organ development.


Subject(s)
Aorta/embryology , Embryonic Induction , Endoderm/physiology , Endothelium, Vascular/physiology , Islets of Langerhans/embryology , Pancreas/embryology , Animals , Aorta/cytology , Aorta/physiology , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Culture Techniques , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Eye Proteins , Gastric Mucosa/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Insulin/biosynthesis , Insulin/genetics , Islets of Langerhans/blood supply , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lymphokines/biosynthesis , Lymphokines/genetics , Mesoderm/physiology , Mice , Mice, Inbred ICR , Mice, Transgenic , Morphogenesis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors , Pancreas/blood supply , Pancreas/cytology , Repressor Proteins , Signal Transduction , Stomach/blood supply , Stomach/cytology , Stomach/embryology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenopus laevis
14.
EMBO J ; 20(18): 5070-8, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11566872

ABSTRACT

The PrP gene of the host exerts a major influence over the outcome of transmissible spongiform encephalopathy (TSE) disease, but the mechanism by which this is achieved is not understood. We have introduced a specific mutation into the endogenous murine PrP gene using gene targeting to produce transgenic mice with a single amino acid alteration (proline to leucine) at amino acid position 101 in their PrP protein (P101L). The effect of this alteration on incubation time, targeting and PrP(Sc) formation has been studied in TSE-infected animals. Transgenic mice carrying the P101L mutation in PrP have remarkable differences in incubation time and targeting of central nervous system pathology compared with wild-type littermates, following inoculation with infectivity from human, hamster, sheep and murine sources. This single mutation can alter incubation time across three species barriers in a strain-dependent manner. These findings suggest a critical role for the structurally 'flexible' region of PrP in agent replication and targeting of TSE pathology.


Subject(s)
Creutzfeldt-Jakob Syndrome/etiology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Prion Diseases/etiology , Prions/genetics , Prions/pharmacology , Scrapie/etiology , Amino Acid Sequence , Animals , Brain/pathology , Creutzfeldt-Jakob Syndrome/pathology , Cricetinae , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Point Mutation , PrPSc Proteins/biosynthesis , Prion Proteins , Recombinant Proteins , Scrapie/metabolism , Sequence Homology, Amino Acid , Sheep , Species Specificity , Time Factors
15.
Mol Cell Endocrinol ; 177(1-2): 117-24, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11377827

ABSTRACT

Using DNA constructs containing regulatory sequences of the zebrafish Pdx-1 and insulin genes, germline transgenic zebrafish expressing the green fluorescent protein (GFP) reporter gene in the pancreas were generated. For both constructs, the GFP expression patterns in transgenic embryos were consistent with the mRNA expression patterns detected by RNA in situ hybridization. A deletion promoter analysis revealed that positive and negative cis-acting elements were involved in regulation of insulin gene expression. Three-dimensional reconstructions imaged from living embryos using two-photon laser-scanning microscopy (TPLSM) demonstrated that the zebrafish pancreas is formed from a single dorsal pancreatic cell mass. This is in contrast to mammals where the pancreas derives from both dorsal and ventral anlage. These transgenic fish should be useful for in vivo studies of factors involved in specifying and regulating pancreatic development and function.


Subject(s)
Homeodomain Proteins , Pancreas/growth & development , Zebrafish/embryology , 5' Untranslated Regions , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/chemistry , Green Fluorescent Proteins , Insulin/genetics , Insulin/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Organ Specificity , Pancreas/embryology , Time Factors , Trans-Activators/metabolism
16.
Mol Cell Neurosci ; 17(4): 768-75, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11312611

ABSTRACT

Ectopic expression of the doppel (Dpl) protein, a homologue of the prion protein (PrP), was recently associated with cerebellar Purkinje cell degeneration observed in two aging prion protein knock-out (Prnp(0/0)) mouse lines. We investigated the possible role of Dpl in oxidative metabolism. Two Prnp(0/0) mouse lines of similar genetic background were studied. One line expresses Dpl in the brain and displays Dpl-associated cerebellar abnormalities. The other has no elevated expression of Dpl and no cerebellar abnormalities. We observed a correlation between Dpl expression and the induction of both heme oxygenase 1 (HO-1) and nitric oxide synthase systems (nNOS and iNOS). These responses are suggestive of increased oxidative stress in the brains of the Dpl-expressing Prnp(0/0) mice. No induction was observed with Hsp-60, indicating a specific response by the HO/NOS system. We proposed that Dpl expression exacerbates oxidative damage that is antagonistic to the protective function of wild-type PrP.


Subject(s)
Heme Oxygenase (Decyclizing)/genetics , Nitric Oxide Synthase/genetics , Prions/genetics , Prions/metabolism , Purkinje Cells/enzymology , Animals , Chaperonin 60/genetics , GPI-Linked Proteins , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1 , Lipid Peroxidation/physiology , Membrane Proteins , Mice , Mice, Knockout , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitrites/metabolism , Oxidative Stress/physiology
17.
Transgenic Res ; 10(2): 133-42, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11305360

ABSTRACT

Gene amplification is widely used for the production of pharmaceuticals and therapeutics in situations where a mammalian system is essential to synthesise a fully active product. Current gene amplification systems require multiple rounds of selection, often with high concentrations of toxic chemicals, to achieve the highest levels of gene amplification. The use of these systems has not been demonstrated in specialised mammalian cells, such as embryonic-stem cells, which can be used to generate transgenic animals. Thus, it has not yet proved possible to produce transgenic animals containing amplified copies of a gene of interest, with the potential to synthesise large amounts of a valuable gene product. We have developed a new amplification system, based around vectors encoding a partially disabled hypoxanthine phosphoribosyltransferase (HPRT) minigene, which can achieve greater than 1000-fold amplification of HPRT and the human growth hormone gene in a single step in Chinese hamster-lung cells. The amplification system also works in mouse embryonic-stem cells and we have used it to produce mice which express 30-fold higher levels of human protein C in milk than obtained with conventional transgenesis using the same protein C construct. This system should also be applicable to large animal transgenics produced by nuclear transfer from cultured cell lines.


Subject(s)
Animals, Genetically Modified/genetics , Gene Amplification , Animals , Cells, Cultured , Cricetinae , Cricetulus , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Guinea Pigs , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Milk/chemistry , Protein C/analysis , RNA, Messenger/genetics , Stem Cells/enzymology
18.
Genes Dev ; 15(4): 444-54, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11230152

ABSTRACT

The mechanisms by which the epithelium of the digestive tract and its associated glands are specified are largely unknown. One clue is that several transcription factors are expressed in specific regions of the endoderm prior to and during organogenesis. Pdx-1, for example, is expressed in the duodenum and pancreas and Pdx-1 inactivation results in an arrest of pancreatic development after buds formation. Similarly, ngn3 is transiently expressed in the developing pancreas and a knockout results in the absence of endocrine cells. This paper focuses on the question of whether these and other transcription factors, known to be necessary for pancreatic development, are also sufficient to drive a program of pancreatic organogenesis. Using in ovo electroporation of chick embryos, we show that ectopic expression of Pdx-1 or ngn3 causes cells to bud out of the epithelium like pancreatic progenitors. The Pdx-1-expressing cells extinguish markers for other nonpancreatic regions of the endoderm and initiate, but do not complete, pancreatic cytodifferentiation. Ectopic expression of ngn3 is sufficient to turn endodermal cells of any region into endocrine cells that form islets expressing glucagon and somatostatin in the mesenchyme. The results suggest that simple gene combinations could be used in stem cells to achieve specific endodermal tissue differentiation.


Subject(s)
Genes, Regulator , Homeodomain Proteins , Pancreas/embryology , Animals , Base Sequence , Body Patterning , Cell Differentiation , Chick Embryo , DNA Primers , Digestive System/embryology , Digestive System/metabolism , Endoderm , Gene Expression Regulation, Developmental , Glucagon/metabolism , Immunohistochemistry , In Situ Hybridization , Nerve Tissue Proteins/physiology , Pancreas/cytology , Pancreas/metabolism , Somatostatin/metabolism , Trans-Activators/genetics
19.
Carcinogenesis ; 22(3): 403-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238179

ABSTRACT

The nucleotide excision repair pathway has evolved to deal with UV light-induced DNA damage. Individuals with the rare inherited nucleotide excision repair deficiency disease xeroderma pigmentosum have a 1000-fold increased incidence of skin cancer. We are interested in the possibility that more subtle changes in nucleotide excision repair genes, resulting in either a reduced capacity for repair or in altered interactions between repair proteins and components of the cell cycle control machinery, might constitute important genetic risk factors for the development of skin cancer in the general population. To investigate this hypothesis we have compared the frequency of polymorphisms in exons 6, 22 and 23 of the XPD gene in melanoma patients and a control group. For each of these two allele polymorphisms one of the alleles was over-represented in the melanoma group and there was a significant association with melanoma. Importantly, this association did not extend to markers immediately flanking the XPD gene, thus providing evidence that XPD gene polymorphisms might predispose to melanoma in the general population. There is a report that one of the polymorphic XPD alleles (exon 23 Lys), which is over-represented in the melanoma group, has reduced repair proficiency and we discuss the possibility that this is the causal change to the XPD gene that predisposes to melanoma.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Genetic Predisposition to Disease , Melanoma/genetics , Polymorphism, Genetic , Proteins/genetics , Transcription Factors , Alleles , Base Sequence , DNA Primers , Genetic Linkage , Humans , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Xeroderma Pigmentosum Group D Protein
20.
Proc Natl Acad Sci U S A ; 97(21): 11307-12, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11027332

ABSTRACT

Human embryonic stem (ES) cells are pluripotent cells derived from the inner cell mass of in vitro fertilized human blastocysts. We examined the potential of eight growth factors [basic fibroblast growth factor (bFGF), transforming growth factor beta1 (TGF-beta1), activin-A, bone morphogenic protein 4 (BMP-4), hepatocyte growth factor (HGF), epidermal growth factor (EGF), beta nerve growth factor (betaNGF), and retinoic acid] to direct the differentiation of human ES-derived cells in vitro. We show that human ES cells that have initiated development as aggregates (embryoid bodies) express a receptor for each of these factors, and that their effects are evident by differentiation into cells with different epithelial or mesenchymal morphologies. Differentiation of the cells was assayed by expression of 24 cell-specific molecular markers that cover all embryonic germ layers and 11 different tissues. Each growth factor has a unique effect that may result from directed differentiation and/or cell selection, and we can divide the overall effects of the factors into three categories: growth factors (Activin-A and TGFbeta1) that mainly induce mesodermal cells; factors (retinoic acid, EGF, BMP-4, and bFGF) that activate ectodermal and mesodermal markers; and factors (NGF and HGF) that allow differentiation into the three embryonic germ layers, including endoderm. None of the growth factors directs differentiation exclusively to one cell type. This analysis sets the stage for directing differentiation of human ES cells in culture and indicates that multiple human cell types may be enriched in vitro by specific factors.


Subject(s)
Cell Differentiation/physiology , Embryo, Mammalian/cytology , Growth Substances/physiology , Stem Cells/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...