Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Aust Orthod J ; 32(2): 175-183, 2016 Nov.
Article in English | MEDLINE | ID: mdl-29509342

ABSTRACT

INTRODUCTION: Neural crest cells make up a transient migratory population of cells found in all vertebrate embryos. Great advances have been made over the past 20 years in clarifying the molecular basis of neural crest induction and, although much still remains unclear, it appears that it is a process involving several factors acting at different stages of embryogenesis. In the future, an understanding of the precise mechanisms involved in orofacial development, even at the earliest stages, may well be of use to all clinicians interested in the management of these tissues. AIM: The present study was designed to determine if the early addition of noggin (a bone morphogenetic protein lBMP) antagonist) and/or the late addition of BMP4 would increase the expression of the transcription factors: Msx-1, Snail, Slug and Pax-7. METHOD: This involved an assessment of the effects of early addition ( Days 0 to 3) of noggin and/or the late addition ( Days 4 to 7) of BMP4 on2the expression of the neural crest markers by human embryonic stem cells, co-cultured for eight days on a feeder layer of mouse PA6 cells. RESULTS AND CONCLUSIONS: The expression of the neural crest markers Pax-7, Msx-1, Slug, and Snail by human embryonic stem cells is likely to be affected by the addition of noggin and BMP4. Not all of these effects will necessarily be significant. The late addition of BMP4 is likely to significantly increase the expression of Pax-7 by human embryonic stem cells (hESCs), when compared with the effects of co-culturing with stromal cell-derived inducing activity, alone. The early addition of noggin and the late addition of BMP4 are likely to significantly increase the expression of Msx-1 by hESCs, when compared with the late addition of BMP4, alone. The hESC results support those from animal ESC studies that the late addition of BMP4, especially, may result in the differentiation of neural crest precursors.


Subject(s)
Human Embryonic Stem Cells/cytology , Neural Crest/cytology , Animals , Bone Morphogenetic Protein 4/pharmacology , Carrier Proteins/pharmacology , Cells, Cultured , Humans , MSX1 Transcription Factor/metabolism , Mice , PAX7 Transcription Factor/metabolism , Photography , Polymerase Chain Reaction , Snail Family Transcription Factors/metabolism
2.
Heliyon ; 10(17): e36298, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39263154

ABSTRACT

Purpose: Current monoclonal antibody-based treatment approaches for cutaneous T cell lymphoma (CTCL) rely heavily on the ability to identify a tumor specific target that is essentially absent on normal cells. Herein, we propose tumor associated glycoprotein-72 (TAG-72) as one such target. TAG-72 is a mucin-associated, truncated O-glycan that has been identified as a chimeric antigen receptor (CAR)-T cell target in solid tumor indications. To date, TAG-72 targeting has not been considered in the setting of hematological malignancies. Experimental design: CD3+ cells from patients with CTCL were analyzed for TAG-72 expression by flow cytometry. Immunohistochemistry was used to assess TAG-72 expression in CTCL patient skin lesions and a TAG-72 ELISA was employed to assess soluble TAG-72 (CA 72-4) in patient plasma. TAG-72 CAR transduction was performed on healthy donor (HD) and CTCL T cells and characterized by flow cytometry. In vitro CAR-T cell function was assessed by flow cytometry and xCELLigence® using patient peripheral blood mononuclear cells and proof-of-concept ovarian cancer cell lines. In vivo CAR-T cell function was assessed in a proof-of-concept, TAG-72+ ovarian cancer xenograft mouse model. Results: TAG-72 expression was significantly higher on total CD3+ T cells and CD4+ subsets in CTCL donors across disease stages, compared to that of HDs. TAG-72 was also present in CTCL patient skin lesions, whereas CA 72-4 was detected at low levels in both CTCL patient and HD plasma with no differences between the two groups. In vitro cytotoxicity assays showed that anti-TAG-72 CAR-T cells significantly, and specifically reduced CD3+TAG-72+ expressing CTCL cells, compared to culture with unedited T cells (no CAR). CTCL CAR-T cells had comparable function to HD CAR-T cells in vitro and CAR-T cells derived from CTCL patients eradicated cancer cells in vivo. Conclusion: This study shows the first evidence of TAG-72 as a possible target for the treatment of CTCL.

3.
Nature ; 450(7169): E12-4, 2007 Nov 22.
Article in English | MEDLINE | ID: mdl-18004280

ABSTRACT

Somatic cell nuclear transfer (SCNT) into enucleated oocytes has emerged as a technique that can be used to derive mouse embryonic stem cell lines with defined genotypes. In this issue Byrne et al. report the derivation of two SCNT Rhesus macaca male stem cell lines designated CRES-1 and CRES-2. Molecular studies detailed in their paper provides supporting evidence that the chromosome complement of CRES-1 and CRES-2 was genetically identical to the male cell donor nucleus and that the mitochondrial DNA originated from different recipient oocytes. In this validation paper, we independently confirm that both stem cell lines were indeed derived by SCNT.


Subject(s)
Macaca mulatta/genetics , Nuclear Transfer Techniques , Pluripotent Stem Cells/metabolism , Animals , Cell Line , Female , Genetic Markers/genetics , Genotype , Humans , Male , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results
4.
Stem Cells ; 29(3): 462-73, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21425409

ABSTRACT

We have used homologous recombination in human embryonic stem cells (hESCs) to insert sequences encoding green fluorescent protein (GFP) into the NKX2.1 locus, a gene required for normal development of the basal forebrain. Generation of NKX2.1-GFP(+) cells was dependent on the concentration, timing, and duration of retinoic acid treatment during differentiation. NKX2.1-GFP(+) progenitors expressed genes characteristic of the basal forebrain, including SHH, DLX1, LHX6, and OLIG2. Time course analysis revealed that NKX2.1-GFP(+) cells could upregulate FOXG1 expression, implying the existence of a novel pathway for the generation of telencephalic neural derivatives. Further maturation of NKX2.1-GFP(+) cells gave rise to γ-aminobutyric acid-, tyrosine hydroxylase-, and somatostatin-expressing neurons as well as to platelet-derived growth factor receptor α-positive oligodendrocyte precursors. These studies highlight the diversity of cell types that can be generated from human NKX2.1(+) progenitors and demonstrate the utility of NKX2.1(GFP/w) hESCs for investigating human forebrain development and neuronal differentiation.


Subject(s)
Cell Lineage/genetics , Cell Tracking/methods , Embryonic Stem Cells/metabolism , Nuclear Proteins/genetics , Prosencephalon/embryology , Transcription Factors/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Flow Cytometry/methods , Genes, Reporter , Humans , Mice , Mice, Transgenic , Molecular Targeted Therapy/methods , Neurogenesis/genetics , Neurogenesis/physiology , Nuclear Proteins/metabolism , Prosencephalon/cytology , Prosencephalon/physiology , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism
5.
Front Immunol ; 13: 968395, 2022.
Article in English | MEDLINE | ID: mdl-36059451

ABSTRACT

Chimeric antigen receptor (CAR)-T therapy has demonstrated remarkable outcomes for B cell malignancies, however, its application for T cell lymphoma, particularly cutaneous T cell lymphoma (CTCL), has been limited. Barriers to effective CAR-T cell therapy in treating CTCL include T cell aplasia in autologous transplants, CAR-T product contamination with leukemic T cells, CAR-T fratricide (when the target antigen is present on normal T cells), and tumor heterogeneity. To address these critical challenges, innovative CAR engineering by targeting multiple antigens to strike a balance between efficacy and safety of the therapy is necessary. In this review, we discuss the current obstacles to CAR-T cell therapy and highlight potential targets in treating CTCL. Looking forward, we propose strategies to develop more powerful dual CARs that are advancing towards the clinic in CTCL therapy.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Receptors, Chimeric Antigen , Skin Neoplasms , Humans , Immunotherapy, Adoptive/adverse effects , Lymphoma, T-Cell, Cutaneous/therapy , Receptors, Chimeric Antigen/genetics , Skin Neoplasms/therapy , T-Lymphocytes
6.
Mol Ther Oncolytics ; 20: 325-341, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33614914

ABSTRACT

Chimeric antigen receptor (CAR) T cells have revolutionized blood cancer immunotherapy; however, their efficacy against solid tumors has been limited. A common mechanism of tumor escape from single target therapies is downregulation or mutational loss of the nominal epitope. Targeting multiple antigens may thus improve the effectiveness of CAR immunotherapies. We generated dual CAR-T cells targeting two tumor antigens: TAG-72 (tumor-associated glycoprotein 72) and CD47. TAG-72 is a pan-adenocarcinoma oncofetal antigen, highly expressed in ovarian cancers, with increased expression linked to disease progression. CD47 is ubiquitously overexpressed in multiple tumor types, including ovarian cancer; it is a macrophage "don't eat me" signal. However, CD47 is also expressed on many normal cells. To avoid this component of the dual CAR-T cells killing healthy tissue, we designed a truncated CD47 CAR devoid of intracellular signaling domains. The CD47 CAR facilitates binding to CD47+ cells, increasing the prospect of TAG-72+ cell elimination via the TAG-72 CAR. Furthermore, we could reduce the damage to normal tissue by monomerizing the CD47 CAR. Our results indicate that the co-expression of the TAG-72 CAR and the CD47-truncated monomer CAR on T cells could be an effective, dual CAR-T cell strategy for ovarian cancer, also applicable to other adenocarcinomas.

7.
Reprod Biomed Online ; 19(1): 106-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19573298

ABSTRACT

Huntington disease (HD) is an incurable late-onset neurodegenerative disorder caused by a CAG repeat expansion in exon 1 of the HD gene (HTT). The major hallmark of disease pathology is neurodegeneration in the brain. Currently, there are no useful in-vitro human models of HD. Recently, two human embryonic stem cell (hESC) lines carrying partial (CAG(37)) and fully (CAG(51)) penetrant mutant alleles have been derived from affected IVF embryos identified following preimplantation genetic diagnosis (PGD). Fluorescence polymerase chain reaction (F-PCR) and Genescan analysis confirmed the original embryonic HD genotypes. Reverse transcription PCR (RT-PCR) analysis confirmed the expression of mutant transcripts and western blot analysis demonstrated expression of mutant huntingtin protein (HTT). After treatment with noggin, HD hESC formed neurospheres, which could be further differentiated into cells susceptible to neurodegeneration in HD, namely primary neurones and astrocytes. Small pool PCR analysis of neurosphere cells revealed instability of disease-length CAG repeats following differentiation. The presence of active HTT genes, neural differentiation capabilities and evidence of CAG repeat instability indicates these HD hESC lines may serve as valuable in-vitro human models of HD to better understand the mechanisms of neurodegeneration in patients, and for drug screening to identify new therapies for human clinical trials.


Subject(s)
Embryonic Stem Cells/cytology , Huntington Disease/pathology , Models, Biological , Blotting, Western , Cell Differentiation , Cell Line , Humans , Huntingtin Protein , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trinucleotide Repeats
8.
Hum Reprod ; 23(8): 1748-59, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18477572

ABSTRACT

BACKGROUND: Currently there are no markers fully predictive of developmental competence of human IVF embryos. The present study investigated a novel strategy involving blastocyst biopsy and DNA fingerprinting to link developmental competence with gene expression patterns. METHODS: Patient's blastocysts were biopsied to remove 8-20 trophectoderm (TE) cells for molecular analysis prior to transfer. Biopsy samples were amplified and gene expression was evaluated using microarrays. Sibling TE biopsies and cells from resulting offspring were subjected to DNA fingerprinting to identify which blastocyst(s) in the transfer cohort developed to term. RESULTS: Blastocyst biopsy did not appear to impair developmental competence. Comparative microarray analysis of cDNA from pooled 'viable' and 'non-viable' TE samples identified over 7000 transcripts expressed exclusively in 'viable' blastocysts. The most significant of these included transcripts involved in cell adhesion and cell communication, key processes that have been associated with mammalian implantation. DNA fingerprinting of three cohorts of sibling blastocysts identified those blastocyst(s) that produced term pregnancies. CONCLUSIONS: The combination of blastocyst biopsy, microarray gene expression profiling and DNA fingerprinting is a powerful tool to identify diagnostic markers of competence to develop to term. This strategy may be used to develop a rapid diagnostic assay or for refining existing criteria for the selection of the single most viable blastocyst among a cohort developing in vitro.


Subject(s)
Blastocyst/physiology , Embryonic Development/physiology , Fertilization in Vitro/methods , Biopsy , DNA Fingerprinting , Embryo Culture Techniques , Embryo Implantation , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Pregnancy
9.
Hum Reprod ; 23(5): 1138-44, 2008 May.
Article in English | MEDLINE | ID: mdl-18346995

ABSTRACT

BACKGROUND: Immature human oocytes matured in vitro, particularly those from gonadotrophin stimulated ovaries, are developmentally incompetent when compared with oocytes matured in vivo. This developmental incompetence has been explained as poor oocyte cytoplasmic maturation without any determination of the likely molecular basis of this observation. METHODS: Replicate whole human genome arrays were generated for immature and mature oocytes (matured in vivo and in vitro, prior to exposure to sperm) recovered from women undertaking gonadotrophin treatment for assisted reproduction. RESULTS: More than 2000 genes were identified as expressed at more than 2-fold higher levels in oocytes matured in vitro than those matured in vivo (P < 0.05, range 4.98 x 10(-2) -2.22 x 10(-4)) and 162 of these are expressed at 10-fold or greater levels (P < 0.05, range 4.98 x 10(-2)-1.38 x 10(-3)). Many of these genes are involved in transcription, the cell cycle and its regulation, transport and cellular protein metabolism. CONCLUSIONS: Global gene expression profiling using microarrays and bioinformatics analysis has provided a molecular basis for differences in the developmental competence of oocytes matured in vitro compared with in vivo. The over-abundance of transcripts identified in immature germinal vesicle stage oocytes recovered from gonadotrophin stimulated cycles and matured in vitro is probably due to dysregulation in either gene transcription or post-transcriptional modification of genes. Either mechanism would result in an incorrect temporal utilization of genes which may culminate in developmental incompetence of any embryos derived from these oocytes.


Subject(s)
Gene Expression Profiling , Oocytes/physiology , Female , Genome, Human/physiology , Humans , In Vitro Techniques , Oligonucleotide Array Sequence Analysis , Superovulation
10.
Reprod Fertil Dev ; 19(6): 740-7, 2007.
Article in English | MEDLINE | ID: mdl-17714628

ABSTRACT

Reproductive technologies have made impressive advances since the 1950s owing to the development of new and innovative technologies. Most of these advances were driven largely by commercial opportunities and the potential improvement of farm livestock production and human health. Companion animals live long and healthy lives and the greatest expense for pet owners are services related to veterinary care and healthcare products. The recent development of embryonic stem cell and nuclear transfer technology in primates and mice has enabled the production of individual specific embryonic stem cell lines in a number of species for potential cell-replacement therapy. Stem cell technology is a fast-developing area in companion animals because many of the diseases and musculoskeletal injuries of cats, dogs and horses are similar to those in humans. Nuclear transfer-derived stem cells may also be selected and directed into differentiation pathways leading to the production of specific cell types, tissues and, eventually, even organs for research and transplantaton. Furthermore, investigations into the treatment of inherited or acquired pathologies have been performed mainly in mice. However, mouse models do not always faithfully represent the human disease. Naturally occurring diseases in companion animals can be more ideal as disease models of human genetic and acquired diseases and could help to define the potential therapeutic efficiency and safety of stem cell therapies. In the present review, we focus on the economic implications of companion animals in society, as well as recent biotechnological progress that has been made in horse, dog and cat embryonic stem cell derivation.


Subject(s)
Breeding/methods , Cats , Dogs , Embryonic Stem Cells , Horses , Nuclear Transfer Techniques , Animals , Biotechnology , Disease Models, Animal
11.
Fertil Steril ; 108(4): 620-627.e4, 2017 10.
Article in English | MEDLINE | ID: mdl-28863935

ABSTRACT

OBJECTIVE: To develop and validate a new strategy to distinguish between balanced/euploid carrier and noncarrier embryos in preimplantation genetic diagnosis (PGD) cycles for reciprocal translocations and to successfully achieve a live birth after selective transfer of a noncarrier embryo. DESIGN: Retrospective and prospective study. SETTING: In vitro fertilization (IVF) units. PATIENT(S): Eleven patients undergoing mate pair sequencing for identification of translocation breakpoints, followed by clinical PGD cycles. INTERVENTION(S): Embryo biopsy with 24-chromosome testing to determine carrier status of balanced/euploid embryos. MAIN OUTCOME MEASURE(S): Definition of translocation breakpoints and polymerase chain reaction (PCR) diagnostic primers, correct diagnosis of euploid embryos for carrier status, and a live birth with a normal karyotype after transfer of a noncarrier embryo. RESULT(S): In 9 of 11 patients (82%), translocation breakpoints were successfully identified. In four patients with a term PGD pregnancy established with a balanced/euploid embryo of unknown carrier status, the correct carrier status was retrospectively determined, matching with the cytogenetic karyotype of the resulting newborns. In a prospective PGD cycle undertaken by a patient with a 46,XY,t(7;14)(q22;q24.3) translocation, the four balanced/euploid embryos identified comprised three carriers and one noncarrier. Transfer of the noncarrier embryo resulted in birth of a healthy girl who was subsequently confirmed with a normal 46,XX karyotype. CONCLUSION(S): The combination of mate pair sequencing and PCR breakpoint analysis of balanced reciprocal translocation derivatives is a novel, reliable, and accurate strategy for distinguishing between carrier and noncarrier balanced/euploid embryos. The method has potential application in clinical PGD cycles for patients with reciprocal translocations or other structural rearrangements.


Subject(s)
Embryo Transfer/methods , Fertilization in Vitro , Genetic Carrier Screening/methods , Preimplantation Diagnosis/methods , Translocation, Genetic , Adult , Female , Fertilization in Vitro/methods , Humans , Infant, Newborn , Karyotyping , Male , Ploidies , Polymerase Chain Reaction/methods , Pregnancy , Prenatal Diagnosis/methods , Retrospective Studies
13.
Biomaterials ; 27(4): 615-22, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16099498

ABSTRACT

Apatites play a crucial role in the body and have been used extensively in biomedical implants. The influence on stem cell behaviour is not known and so this study will explore whether sintered carbonated apatites are favourable for propagation of stem cells. Different weight substitutions of carbonated apatite, specifically 2.5 wt% (2.5 wt%CAP) and 5 wt% (5 wt%CAP), were sintered and characterised prior to the investigation of their potential as a matrix for the support of mouse embryonic stem (ES) cells. Characterisation of the apatites included elemental analysis, X-ray diffraction, surface roughness, specific surface area, density, and solubility. The ability of carbonated apatite to support mouse ES cell colonisation and maintenance in the presence of leukaemia inhibitory factor was determined by an enumeration of live versus dead cells within a population, and immunoreactivity to Oct4, a transcription factor and stem cell marker, following growth on each matrix. It was found that while both compositions allowed for the colonisation of mouse ES cells, the cells were not maintained in an undifferentiated state, as evidenced by a reduction in the number of cells staining positive for Oct4 expression. This study shows that an increase in carbonate content within sintered apatites leads to a higher cell number, a desired aspect for stem cells to populate scaffolds intended for tissue engineering. This study presents carbonated apatites as a suitable matrix for the initial colonisation and differentiation of ES cells for tissue engineering applications.


Subject(s)
Apatites/chemistry , Biocompatible Materials/chemistry , Carbon/chemistry , Cell Culture Techniques/methods , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering/methods , Animals , Cell Count , Cell Differentiation , Cell Proliferation , Cell Size , Cell Survival , Cells, Cultured , Materials Testing , Mice , Stem Cell Transplantation/methods
14.
Microsc Res Tech ; 69(6): 396-407, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16718650

ABSTRACT

The fine structure of human oogonia and growing oocytes has been reviewed in fetal and adult ovaries. Preovulatory maturation and the ultrastructure of stimulated oocytes from the germinal vesicle (GV) stage to metaphase II (MII) stage are also documented. Oogonia have large nuclei, scanty cytoplasm with complex mitochondria. During folliculogenesis, follicle cell processes establish desmosomes and deep gap junctions at the surface of growing oocytes, which are retracted during the final stages of maturation. The zona pellucida is secreted in secondary follicles. Growing oocytes have mitochondria, Golgi, rough endoplasmic reticulum (RER), ribosomes, lysosomes, and lipofuscin bodies, often associated with Balbiani bodies and have nuclei with reticulated nucleoli. Oocytes from antral follicles show numerous surface microvilli and cortical granules (CGs) separated from the oolemma by a band of microfilaments. The CGs are evidently secreted by Golgi membranes. The GV oocytes have peripheral Golgi complexes associated with a single layer of CGs close to the oolemma. They have many lysosomes, and nuclei with dense compact nucleoli. GV breakdown occurs by disorganization of the nuclear envelope and the oocyte enters a transient metaphase I followed by MII, when it is arrested and ovulated. Maturation of oocytes in vitro follows the same pattern of meiosis seen in preovulatory oocytes. The general organization of the human oocyte conforms to that of most other mammals but has some unique features. The MII oocyte has the basic cellular organelles such as mitochondria, smooth endoplasmic reticulum, microfilaments, and microtubules, while Golgi, RER, lysosomes, multivesicular, residual and lipofuscin bodies are very rare. It neither has yolk nor lipid inclusions. Its surface has few microvilli, and 1-3 layers of CGs, aligned beneath the oolemma. Special reference has been made to the reduction and inactivation of the maternal centrosome during oogenesis. The MII spindle, often oriented perpendicular to the oocyte surface, is barrel-shaped, anastral and lacks centrioles. Osmiophilic centrosomes are not demonstrable in human eggs, since the maternal centrosome is nonfunctional. However, oogonia and growing oocytes have typical centrioles, similar to those of somatic cells. The sperm centrosome activates the egg and organizes the sperm aster and mitotic spindles of the embryo, after fertilization.


Subject(s)
Centrosome/physiology , Oocytes , Oogonia , Female , Humans , Oocytes/physiology , Oocytes/ultrastructure , Oogonia/cytology , Oogonia/ultrastructure , Organelles/ultrastructure , Ovarian Follicle/cytology , Ovarian Follicle/ultrastructure , Ovary/cytology , Ovary/ultrastructure , Ovum/cytology , Ovum/ultrastructure
15.
Methods Mol Biol ; 348: 319-32, 2006.
Article in English | MEDLINE | ID: mdl-16988390

ABSTRACT

The birth of viable offspring from somatic cell nuclear transfer (SCNT) in mammals caused a major re-examination of the understanding of the commitment of cells to specific tissue lineages during differentiation. The questions of whether cells undergo dedifferentiation or transdifferentiation during the development of offspring and how these changes are controlled is a source of ongoing debate that is yet to be resolved. Irrespective of the outcome of this debate, it is clear that cloning using SCNT has a place and purpose in the future of research and animal breeding. The future uses of SCNT could include the production of transgenic mice, the production of transgenic livestock and assisting with the re-establishment of endangered species. Human medicine also would benefit from future use of SCNT because it would allow the production of patient-specific embryonic stem cells.


Subject(s)
Animals, Genetically Modified , Cell Nucleus/physiology , Cloning, Organism/statistics & numerical data , Cloning, Organism/trends , Epigenesis, Genetic , Animals , Animals, Domestic/genetics , Animals, Wild/genetics , Cloning, Organism/veterinary , Embryo Culture Techniques , Embryo Transfer/veterinary , Humans , Models, Biological , Nuclear Transfer Techniques/veterinary , Stem Cells/physiology
16.
Stem Cell Rev ; 2(4): 265-76, 2006.
Article in English | MEDLINE | ID: mdl-17848713

ABSTRACT

Human therapeutic cloning or nuclear transfer stem cells (NTSC) to produce patient-specific stem cells, holds considerable promise in the field of regenerative medicine. The recent withdrawal of the only scientific publications claiming the successful generation of NTSC lines afford an opportunity to review the available research in mammalian reproductive somatic cell nuclear transfer (SCNT) with the goal of progressing human NTSC. The process of SCNT is prone to epigenetic abnormalities that contribute to very low success rates. Although there are high mortality rates in some species of cloned animals, most surviving clones have been shown to have normal phenotypic and physiological characteristics and to produce healthy offspring. This technology has been applied to an increasing number of mammals for utility in research, agriculture, conservation, and biomedicine. In contrast, attempts at SCNT to produce human embryonic stem cells (hESCs) have been disappointing. Only one group has published reliable evidence of success in deriving a cloned human blastocyst, using an undifferentiated hESC donor cell, and it failed to develop into a hESC line. When optimal conditions are present, it appears that in vitro development of cloned and parthenogenetic embryos, both of which may be utilized to produce hESCs, may be similar to in vitro fertilized embryos. The derivation of ESC lines from cloned embryos is substantially more efficient than the production of viable offspring. This review summarizes developments in mammalian reproductive cloning, cell-to-cell fusion alternatives, and strategies for oocyte procurement that may provide important clues facilitating progress in human therapeutic cloning leading to the successful application of cell-based therapies utilizing autologous hESC lines.


Subject(s)
Cloning, Organism , Embryonic Stem Cells/physiology , Nuclear Transfer Techniques , Animals , Cell Differentiation , Cellular Reprogramming , Embryo, Mammalian/physiology , Humans , Hybrid Cells/physiology , Oocytes/physiology
17.
Stem Cell Rev ; 2(4): 277-87, 2006.
Article in English | MEDLINE | ID: mdl-17848714

ABSTRACT

Nuclear transfer (NT) experiments in mammals have demonstrated that adult cells are genetically equivalent to early embryonic cells and the reversal of the differentiated state of a cell to another that has characteristics of the undifferentiated embryonic state can be defined as nuclear reprogramming. The feasibility of interspecies somatic cell NT (iSCNT) has been demonstrated by blastocyst formation and the production of offspring in a number of studies. Embryo and oocyte availability is a major limiting factor in conducting NT to obtain, blastocysts for both reproductive NT studies in genetically endangered animals and in embryonic stem cell derivation for species such as the horse and human. One approach to generate new embryonic stem cells in human as disease models, or in species where embryos and oocytes are not widely available, is to use oocytes from another species. Utilization of oocytes for recipient cytoplasts from other species that are accessible and abundant, such as the cow and rabbit, would greatly benefit ongoing research on reprogramming and stem cell sciences. The use of iSCNT is an exciting possibility for species with limited availability of oocytes as well as for endangered or exotic species where assisted reproduction is needed. However, the mechanisms involved in nuclear reprogramming by the oocyte are still unknown and the extent of the "universality" of ooplasmic reprogramming of development remains under investigation.


Subject(s)
Chimera , Hybrid Cells/physiology , Nuclear Transfer Techniques , Animals , Cattle , Cell Nucleus/physiology , DNA, Mitochondrial , Horses , Humans , Mice , Mitochondria/metabolism , Oocytes/physiology
18.
Stem Cell Rev ; 2(4): 331-40, 2006.
Article in English | MEDLINE | ID: mdl-17848720

ABSTRACT

Embryonic stem cell (ESC) technology should enable the generation of specific cell types for the study and treatment of human diseases. Therapeutic cloning provides a way to generate ESCs genetically matched to diseased individuals through nuclear reprogramming of the somatic genome. However, practical and ethical limitations associated with therapeutic cloning are calling for the development of oocyte- and-embryo-free alternatives for obtaining of autologous pluripotent cells for transplantation therapy. An alternative approach to reprogram the somatic genome involves fusion between somatic and pluripotent cells. Potential fusion partners with reprogramming activities include embryonal carcinoma cells, embryonic germ cells, and ESCs. Experimental evidence is now available, which demonstrates that mouse and human somatic cells can be reprogrammed by fusion to form pluripotent hybrid cells. Recent progress infusion-based reprogramming is reviewed with reference to the developmental potency of hybrid cells as well as genetic and epigenetic correlates of reprogramming. However, hybrid cells lack therapeutic potential because of their abnormal ploidy and the presence of nonautologous genes from the pluripotent parent. We discuss the potential of fusion-based reprogramming for the generation of diploid, autologous pluripotent cells using two alternative routes: the enucleation of ESCs and the fusion of such cytoplasts to somatic cell karyoplasts or intact somatic cells, and the selective elimination of the pluripotent genome following fusion to the somatic partner. Finally, these approaches are discussed in the light of recent progress showing that overexpression of embryonic transcription factors can restore a state of pluripotency to somatic cells.


Subject(s)
Cellular Reprogramming , Hybrid Cells/physiology , Nuclear Transfer Techniques , Pluripotent Stem Cells/physiology , Animals , Cell Fusion , Humans
19.
Reprod Fertil Dev ; 17(8): 799-808, 2005.
Article in English | MEDLINE | ID: mdl-16476207

ABSTRACT

Attempts to support survival of mammalian embryos after hatching have met with limited success, although some mouse studies have reported growth at the post-implantation stage. The aim of the present research was to establish and characterise an in vitro culture system that could support extended growth and differentiation of bovine embryos. Abattoir-derived oocytes were matured and fertilised in vitro. Presumptive zygotes were cultured in modified synthetic oviduct fluid (SOFaaci) medium supplemented with 5% cow serum (CS). On Day 9, single hatched blastocysts (n = 160) were randomly allocated to SOFaaci supplemented with either 5% bovine serum albumin, 5% CS, 5% fetal calf serum (FCS) or SOF only and cultured on a collagen gel substrate for up to 45 days. Embryos were evaluated at various time-points until complete disaggregation or the total disappearance of embryonic cells. Blastocyst viability post hatching was severely compromised in protein-free SOFaaci medium. Addition of FCS generated increased embryonic growth for the longest time period (Day 45) when compared to the other groups. Long-term survival of embryonic cells was observed stereomicroscopically by the proliferation and development of three-dimensional tubular structures to 85% confluence in culture. Haematoxylin and eosin staining of morphological structures obtained from all treatment groups revealed embryos displaying trophoblast, inner cell mass and hypoblast development to varying degrees. Regardless of treatment, extended in vitro culture did not result in development comparable with that described for in vivo embryos. In the present work, however, there was evidence of extended culture of bovine embryos beyond that achieved previously. However, further research is required to identify the exact requirements for extended in vitro culture for bovine embryos.


Subject(s)
Embryo Culture Techniques/methods , Embryo Culture Techniques/veterinary , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Fertilization in Vitro/veterinary , Animals , Cattle , Culture Media/chemistry , Fertilization in Vitro/methods , Histological Techniques/veterinary , Survival Analysis
20.
Reprod Fertil Dev ; 17(5): 573-85, 2005.
Article in English | MEDLINE | ID: mdl-15907283

ABSTRACT

The aim of the present study was to compare the in vitro and in vivo developmental competence of hand-made cloning (HMC) embryos with the conventional nuclear transfer (NT) method using five somatic cell lines and in vitro-fertilised (IVF; control) embryos. Modifications to the HMC procedure included fusion efficiency optimisation, effect of cytoplasmic volume and cloned embryo aggregation. The developmental competence of blastocysts from each of the treatment groups and cell lines used was assessed following transfer to 345 recipients. Vitrification was also used to enable management of recipient resources and to assess the susceptibility of membranes to cryopreservation following zona removal. Increasing cytoplasmic volume to 150% or aggregating two embryos improved the blastocyst development rate and increased the total cell number. Although HMC embryo transfers established a significantly higher pregnancy rate on Day 30 than fresh IVF or NT embryo transfers, the overall outcome in terms of cloned live births derived from either fresh or vitrified/thawed HMC or NT embryo transfers across the five cell lines did not differ. The birth and continued survival of clones produced with HMC technology with equivalent efficiency to NT shows that it can be used as an alternative method for the generation of cloned offspring in the bovine.


Subject(s)
Cattle , Cloning, Organism/veterinary , Nuclear Transfer Techniques , Animals , Blastocyst/physiology , Cattle/embryology , Cloning, Organism/methods , Cryopreservation/veterinary , Cytochalasin B/pharmacology , Cytoplasm/ultrastructure , Embryo Culture Techniques/veterinary , Embryo Transfer/veterinary , Embryonic Development , Female , Fertilization in Vitro/veterinary , Oocytes/drug effects , Oocytes/ultrastructure , Pregnancy , Serum Albumin, Bovine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL