Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Nat Commun ; 14(1): 8031, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38052804

RESUMO

Cancer cells inevitably interact with neighboring host tissue-resident cells during the process of metastatic colonization, establishing a metastatic niche to fuel their survival, growth, and invasion. However, the underlying mechanisms in the metastatic niche are yet to be fully elucidated owing to the lack of methodologies for comprehensively studying the mechanisms of cell-cell interactions in the niche. Here, we improve a split green fluorescent protein (GFP)-based genetically encoded system to develop secretory glycosylphosphatidylinositol-anchored reconstitution-activated proteins to highlight intercellular connections (sGRAPHIC) for efficient fluorescent labeling of tissue-resident cells that neighbor on and putatively interact with cancer cells in deep tissues. The sGRAPHIC system enables the isolation of metastatic niche-associated tissue-resident cells for their characterization using a single-cell RNA sequencing platform. We use this sGRAPHIC-leveraged transcriptomic platform to uncover gene expression patterns in metastatic niche-associated hepatocytes in a murine model of liver metastasis. Among the marker genes of metastatic niche-associated hepatocytes, we identify Lgals3, encoding galectin-3, as a potential pro-metastatic factor that accelerates metastatic growth and invasion.


Assuntos
Neoplasias Hepáticas , Humanos , Camundongos , Animais , Neoplasias Hepáticas/metabolismo , Hepatócitos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Matriz Extracelular/metabolismo , Comunicação Celular
2.
Commun Med (Lond) ; 3(1): 56, 2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076593

RESUMO

BACKGROUND: Base editing via CRISPR-Cas9 has garnered attention as a method for correcting disease-specific mutations without causing double-strand breaks, thereby avoiding large deletions and translocations in the host chromosome. However, its reliance on the protospacer adjacent motif (PAM) can limit its use. We aimed to restore a disease mutation in a patient with severe hemophilia B using base editing with SpCas9-NG, a modified Cas9 with the board PAM flexibility. METHODS: We generated induced pluripotent stem cells (iPSCs) from a patient with hemophilia B (c.947T>C; I316T) and established HEK293 cells and knock-in mice expressing the patient's F9 cDNA. We transduced the cytidine base editor (C>T), including the nickase version of Cas9 (wild-type SpCas9 or SpCas9-NG), into the HEK293 cells and knock-in mice through plasmid transfection and an adeno-associated virus vector, respectively. RESULTS: Here we demonstrate the broad PAM flexibility of SpCas9-NG near the mutation site. The base-editing approach using SpCas9-NG but not wild-type SpCas9 successfully converts C to T at the mutation in the iPSCs. Gene-corrected iPSCs differentiate into hepatocyte-like cells in vitro and express substantial levels of F9 mRNA after subrenal capsule transplantation into immunodeficient mice. Additionally, SpCas9-NG-mediated base editing corrects the mutation in both HEK293 cells and knock-in mice, thereby restoring the production of the coagulation factor. CONCLUSION: A base-editing approach utilizing the broad PAM flexibility of SpCas9-NG can provide a solution for the treatment of genetic diseases, including hemophilia B.


In patients with hemophilia B, the blood does not clot properly, leading to excessive bruising and bleeding. Hemophilia B is caused by an error in a gene called coagulation factor IX (F9). To treat patients with hemophilia B, we might be able to use a technology called CRISPR-Cas9 to edit and correct this genetic error, restoring factor IX function and improving clotting. Here, we test a specific CRISPR-Cas9 approach in cells and animals. We show that we are able to correct the genetic error in F9 in cells isolated from a patient with severe hemophilia B. We also show that we can fix the error in mice and that this increases levels of factor IX in the blood of the mice. With further testing, this gene-editing approach may be a viable therapy for patients with hemophilia B or similar genetic disorders.

3.
Methods Mol Biol ; 2637: 213-221, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36773149

RESUMO

Knock-in therapy, in which an insertion site can be controlled, would be more suitable for the treatment of genetic blood disorders as compared to conventional gene therapy with lentivirus vectors that introduce genes into the genome randomly. Recent advancements in genome editing technology have substantially improved the knock-in efficiency, making it a reality. We present the details of a virus-free CRISPR/Cas9-based genome editing method for bona fide mouse hematopoietic stem cells.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Camundongos , Animais , Edição de Genes/métodos , Terapia Genética/métodos , Lentivirus/genética
4.
J Vis Exp ; (169)2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33749676

RESUMO

Pluripotent stem cell-derived cardiomyocytes (PSC-CMs) can be produced from both embryonic and induced pluripotent stem (ES/iPS) cells. These cells provide promising sources for cardiac disease modeling. For cardiomyopathies, sarcomere shortening is one of the standard physiological assessments that are used with adult cardiomyocytes to examine their disease phenotypes. However, the available methods are not appropriate to assess the contractility of PSC-CMs, as these cells have underdeveloped sarcomeres that are invisible under phase-contrast microscopy. To address this issue and to perform sarcomere shortening with PSC-CMs, fluorescent-tagged sarcomere proteins and fluorescent live-imaging were used. Thin Z-lines and an M-line reside at both ends and the center of a sarcomere, respectively. Z-line proteins - α-Actinin (ACTN2), Telethonin (TCAP), and actin-associated LIM protein (PDLIM3) - and one M-line protein - Myomesin-2 (Myom2) - were tagged with fluorescent proteins. These tagged proteins can be expressed from endogenous alleles as knock-ins or from adeno-associated viruses (AAVs). Here, we introduce the methods to differentiate mouse and human pluripotent stem cells to cardiomyocytes, to produce AAVs, and to perform and analyze live-imaging. We also describe the methods for producing polydimethylsiloxane (PDMS) stamps for a patterned culture of PSC-CMs, which facilitates the analysis of sarcomere shortening with fluorescent-tagged proteins. To assess sarcomere shortening, time-lapse images of the beating cells were recorded at a high framerate (50-100 frames per second) under electrical stimulation (0.5-1 Hz). To analyze sarcomere length over the course of cell contraction, the recorded time-lapse images were subjected to SarcOptiM, a plug-in for ImageJ/Fiji. Our strategy provides a simple platform for investigating cardiac disease phenotypes in PSC-CMs.


Assuntos
Corantes Fluorescentes/metabolismo , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/citologia , Sarcômeros/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Dependovirus/metabolismo , Corpos Embrioides/citologia , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Miócitos Cardíacos/citologia , Coloração e Rotulagem , Imagem com Lapso de Tempo
5.
Mol Ther Methods Clin Dev ; 20: 451-462, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33614821

RESUMO

We conducted two lines of genome-editing experiments of mouse hematopoietic stem cells (HSCs) with the clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated protein 9 (Cas9). First, to evaluate the genome-editing efficiency in mouse bona fide HSCs, we knocked out integrin alpha 2b (Itga2b) with Cas9 ribonucleoprotein (Cas9/RNP) and performed serial transplantation in mice. The knockout efficiency was estimated at approximately 15%. Second, giving an example of X-linked severe combined immunodeficiency (X-SCID) as a target genetic disease, we showed a proof-of-concept of universal gene correction, allowing rescue of most of X-SCID mutations, in a completely non-viral setting. We inserted partial cDNA of interleukin-2 receptor gamma chain (Il2rg) into intron 1 of Il2rg via non-homologous end-joining (NHEJ) with Cas9/RNP and a homology-independent targeted integration (HITI)-based construct. Repaired HSCs reconstituted T lymphocytes and thymuses in SCID mice. Our results show that a non-viral genome-editing of HSCs with CRISPR/Cas9 will help cure genetic diseases.

6.
Exp Hematol ; 95: 46-57.e8, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33395577

RESUMO

We report that a sheep fetal liver provides a microenvironment for generating hematopoietic cells with long-term engrafting capacity and multilineage differentiation potential from human induced pluripotent stem cell (iPSC)-derived hemogenic endothelial cells (HEs). Despite the promise of iPSCs for making any cell types, generating hematopoietic stem and progenitor cells (HSPCs) is still a challenge. We hypothesized that the hematopoietic microenvironment, which exists in fetal liver but is lacking in vitro, turns iPSC-HEs into HSPCs. To test this, we transplanted CD45-negative iPSC-HEs into fetal sheep liver, in which HSPCs first grow. Within 2 months, the transplanted cells became CD45 positive and differentiated into multilineage blood cells in the fetal liver. Then, CD45-positive cells translocated to the bone marrow and were maintained there for 3 years with the capability of multilineage differentiation, indicating that hematopoietic cells with long-term engraftment potential were generated. Moreover, human hematopoietic cells were temporally enriched by xenogeneic donor-lymphocyte infusion into the sheep. This study could serve as a foundation to generate HSPCs from iPSCs.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Ovinos/embriologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Microambiente Celular , Ensaio de Unidades Formadoras de Colônias , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Feminino , Técnicas Genéticas , Sobrevivência de Enxerto , Hemangioblastos/citologia , Xenoenxertos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Antígenos Comuns de Leucócito/análise , Fígado/embriologia , Subpopulações de Linfócitos , Gravidez , Especificidade da Espécie
7.
Oncotarget ; 9(5): 6450-6462, 2018 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-29464084

RESUMO

Human induced pluripotent stem cells (hiPSCs) are creating great expectations for regenerative medicine. However, safety strategies must be put in place to guard against teratoma formation after transplantation of hiPSC-derived cells into patients. Recent studies indicate that epigenetic regulators act at the initial step of tumorigenesis. Using gain-of-function and loss-of-function approaches, we show here that the expression and function of lysine-specific demethylase 1 (LSD1) are tightly regulated in hiPSCs, and their deregulation underlies the development of teratomas. Consistent with these results, we demonstrate that an LSD1 inhibitor, S2157, prevented teratoma formation from hiPSCs transplanted into immunodeficient mice. This novel action of LSD1 and the effects of its inhibition potentially allow for the development of new clinical applications and therapeutic strategies using hiPSCs.

8.
Int J Hematol ; 106(5): 631-637, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28687990

RESUMO

We and others have reported that human hematopoietic stem cells (HSCs) are also present in the CD34-negative (CD34-) fraction of human cord blood (CB). Here, we examined the hematopoietic engraftment potential of 13 or 18 lineage-negative (13Lin- or 18Lin-) CD34+/- cells from human CB in mice and sheep. Both 13Lin- and 18Lin- CD34+ cells efficiently engrafted in mice irrespective of transplantation route, be it by tail-vein injection (TVI) or by intra-bone marrow injection (IBMI). These cells also engrafted in sheep after in utero fetal intra-hepatic injection (IHI). In contrast, neither 13Lin- nor 18Lin- CD34- cells engrafted in either mice or sheep when transplanted by regular routes (i.e., TVI and fetal IHI, respectively), although both 13Lin- and 18Lin- CD34- cells engrafted in mice when transplanted by IBMI and exhibited multilineage reconstitution ability. Thus, the homing ability of CD34- HSCs is significantly more limited than that of CD34+ HSCs. As for 18Lin-, CD34- HSCs are characterized by low expression of the tetraspanin CD9, which promotes homing, and high expression of the peptidase CD26, which inhibits homing. This unique expression pattern homing-related molecules on CD34- HSCs could thus explain in part their reduced ability to home to the BM niche.


Assuntos
Dipeptidil Peptidase 4/biossíntese , Regulação da Expressão Gênica/fisiologia , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Tetraspanina 29/biossíntese , Animais , Antígenos CD34 , Feminino , Células-Tronco Hematopoéticas/citologia , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ovinos
9.
Sci Rep ; 7(1): 4159, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28646206

RESUMO

Haemophilia B, a congenital haemorrhagic disease caused by mutations in coagulation factor IX gene (F9), is considered an appropriate target for genome editing technology. Here, we describe treatment strategies for haemophilia B mice using the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 system. Administration of adeno-associated virus (AAV) 8 vector harbouring Staphylococcus aureus Cas9 (SaCas9) and single guide RNA (sgRNA) to wild-type adult mice induced a double-strand break (DSB) at the target site of F9 in hepatocytes, sufficiently developing haemophilia B. Mutation-specific gene editing by simultaneous induction of homology-directed repair (HDR) sufficiently increased FIX levels to correct the disease phenotype. Insertion of F9 cDNA into the intron more efficiently restored haemostasis via both processes of non-homologous end-joining (NHEJ) and HDR following DSB. Notably, these therapies also cured neonate mice with haemophilia, which cannot be achieved with conventional gene therapy with AAV vector. Ongoing haemophilia therapy targeting the antithrombin gene with antisense oligonucleotide could be replaced by SaCas9/sgRNA-expressing AAV8 vector. Our results suggest that CRISPR/Cas9-mediated genome editing using an AAV8 vector provides a flexible approach to induce DSB at target genes in hepatocytes and could be a good strategy for haemophilia gene therapy.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Dependovirus/genética , Edição de Genes , Vetores Genéticos/administração & dosagem , Hemofilia B/terapia , Animais , Animais Recém-Nascidos , Antitrombina III/metabolismo , Sequência de Bases , DNA Complementar/genética , Éxons/genética , Fator IX/metabolismo , Íntrons/genética , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Reparo de DNA por Recombinação/genética
10.
Exp Anim ; 65(4): 345-351, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27333841

RESUMO

Animal models of thrombocytopenia are indispensable for evaluating the in vivo efficacy of hemostatic agents, cryopreserved platelets, and artificial platelets, but no large animal models are available. In this study, we generated a swine model of acute thrombocytopenia with prolonged bleeding times by administering the chemotherapeutic drug busulfan. First, we tested multiple doses of busulfan (4, 6, and 8 mg/kg) in pigs, and found that 6 mg/kg of busulfan is an optimal dose for producing a safe and moderate thrombocytopenia, with a platelet count of less than 30,000/µl. The pigs administered 6 mg/kg of busulfan (n=8) reached half their initial counts at day 7, counts below 30,000/µl at day 12, and their nadirs at day 15 (on average). The minimal platelet count was 14,000/µl. With this dose of busulfan (6 mg/kg), bleeding times were significantly prolonged in addition to the decrease in platelet counts (r=-0.63, P<0.01), while there were no cases of apparent hemorrhage. White blood cell counts were maintained at over 5,000/µl, and there were no infections or other adverse events including anemia or appetite or body weight loss. All pigs were sacrificed on day 16, with subsequent examination showing a significant reduction in cellularity and colony-forming units in the bone marrow, indicating that thrombocytopenia was the result of myelosuppression. In summary, administration with 6 mg/kg of busulfan induces safe and moderate thrombocytopenia with a prolonged bleeding time in swine.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Bussulfano/efeitos adversos , Modelos Animais de Doenças , Trombocitopenia/etiologia , Doença Aguda , Animais , Tempo de Sangramento , Relação Dose-Resposta a Droga , Feminino , Testes Hematológicos , Humanos , Masculino , Suínos , Porco Miniatura
11.
Exp Anim ; 63(4): 475-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25048264

RESUMO

Xenograft models of human hematopoiesis are essential to the study of the engraftment and proliferative potential of human hematopoietic stem cells (HSCs) in vivo. Immunodeficient mice and fetal sheep are often used as xenogeneic recipients because they are immunologically naive. In this study, we transplanted human HSCs into fetal sheep and assessed the long-term engraftment of transplanted human HSCs after birth. Fourteen sheep were used in this study. In 4 fetal sheep, HSCs were transduced with homeo-box B4 (HOXB4) gene before transplantation, which promoted the expansion of HSCs. Another 4 fetal sheep were subjected to non-myeloablative conditioning with busulfan. Seven of these 8 sheep showed successful engraftment of human HSCs (1-3% of colony-forming units) as assessed after the birth of fetal sheep (5 months post-transplantation), although HOXB4-transduced HSCs showed sustained engraftment for up to 40 months. Intact HSCs were transplanted into six non-conditioned fetal sheep, and human colony-forming units were not detected in the sheep after birth. These results suggest that, as compared with mouse models, where the short lifespan of mice limits long-term follow-up of HSC engraftment, the fetal sheep model provides a unique perspective for evaluating long-term engraftment and proliferation of human HSCs.


Assuntos
Proliferação de Células , Feto , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Ovinos/embriologia , Animais , Bussulfano , Feminino , Seguimentos , Xenoenxertos , Proteínas de Homeodomínio , Humanos , Camundongos , Modelos Animais , Gravidez , Fatores de Tempo , Fatores de Transcrição , Transdução Genética , Condicionamento Pré-Transplante
12.
PLoS One ; 9(6): e98319, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24927426

RESUMO

Recent studies have revealed negligible immunogenicity of induced pluripotent stem (iPS) cells in syngeneic mice and in autologous monkeys. Therefore, human iPS cells would not elicit immune responses in the autologous setting. However, given that human leukocyte antigen (HLA)-matched allogeneic iPS cells would likely be used for medical applications, a more faithful model system is needed to reflect HLA-matched allogeneic settings. Here we examined whether iPS cells induce immune responses in the swine leukocyte antigen (SLA)-matched setting. iPS cells were generated from the SLA-defined C1 strain of Clawn miniature swine, which were confirmed to develop teratomas in mice, and transplanted into the testes (n = 4) and ovary (n = 1) of C1 pigs. No teratomas were found in pigs on 47 to 125 days after transplantation. A Mixed lymphocyte reaction revealed that T-cell responses to the transplanted MHC-matched (C1) iPS cells were significantly lower compared to allogeneic cells. The humoral immune responses were also attenuated in the C1-to-C1 setting. More importantly, even MHC-matched iPS cells were susceptible to innate immunity, NK cells and serum complement. iPS cells lacked the expression of SLA class I and sialic acids. The in vitro cytotoxic assay showed that C1 iPS cells were targeted by NK cells and serum complement of C1. In vivo, the C1 iPS cells developed larger teratomas in NK-deficient NOG (T-B-NK-) mice (n = 10) than in NK-competent NOD/SCID (T-B-NK+) mice (n = 8) (p<0.01). In addition, C1 iPS cell failed to form teratomas after incubation with the porcine complement-active serum. Taken together, MHC-matched iPS cells can attenuate cellular and humoral immune responses, but still susceptible to innate immunity in pigs.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Células Matadoras Naturais/imunologia , Complexo Principal de Histocompatibilidade , Porco Miniatura/imunologia , Suínos/imunologia , Teratoma/etiologia , Animais , Células Cultivadas , Feminino , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II , Imunidade Humoral , Imunidade Inata , Células-Tronco Pluripotentes Induzidas/imunologia , Masculino , Camundongos , Camundongos SCID , Ovário/imunologia , Testículo/imunologia , Transplante Autólogo
14.
Stem Cells Dev ; 22(3): 473-82, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22889279

RESUMO

In pluripotent stem cells (PSCs), there are 2 types: naive and primed. Only the naive type has the capacity for producing chimeric offspring. Mouse PSCs are naive, but human PSCs are in the primed state. Previously reported porcine PSCs appear in the primed state. In this study, putative naive porcine-induced pluripotent stem cells (iPSCs) were generated. Porcine embryonic fibroblasts were transduced with retroviral vectors expressing Yamanaka's 4 genes. Emergent colonies were propagated in the presence of porcine leukemia inhibitory factor (pLIF) and forskolin. The cells expressed pluripotency markers and formed embryoid bodies, which gave rise to cell types from all 3 embryonic germ layers. The naive state of the cells was demonstrated by pLIF dependency, 2 active X chromosomes (when female), absent MHC class I expression, and characteristic gene expression profiles. The porcine iPSCs contributed to the in vitro embryonic development (11/24, 45.8%) as assessed by fluorescent markers. They also contributed to the in utero fetal development (11/71, 15.5% at day 23; 1/13, 7.7% at day 65). This is the first demonstration of macroscopic fluorescent chimeras derived from naive-like porcine PSCs, although adult chimeras remain to be produced.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Animais Geneticamente Modificados , Células Cultivadas , Quimera , Metilação de DNA , Corpos Embrioides/metabolismo , Desenvolvimento Embrionário , Feminino , Desenvolvimento Fetal , Células-Tronco Pluripotentes Induzidas/transplante , Mórula/citologia , Fator 3 de Transcrição de Octâmero/genética , Gravidez , Regiões Promotoras Genéticas , Sus scrofa , Transdução Genética , Transgenes , Cromossomo X/genética
15.
Exp Hematol ; 40(6): 436-44, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22306296

RESUMO

In utero transplantation (IUT) of human hematopoietic stem cells has been conducted in sheep, which are used as large animal models of human hematopoietic reconstitution and models for clinical IUT; however, the levels of engraftment have generally been low. Busulfan (BU), a myeloablative agent, is often administered to patients before hematopoietic stem cells transplantation to improve the engraftment. In this study, hematopoietic activity was evaluated in adult sheep after administering BU at different doses. Next, pregnant ewes were administered BU, and dams as well as their fetuses were evaluated, as BU readily crosses the sheep placenta. Then, the BU dose with the desired outcomes was selected and administered to pregnant ewes at 2 or 6 days before performing IUT using human cord blood CD34(+) cells. The engraftment was evaluated in recipients that underwent IUT in the presence or absence of BU. As a result, hematopoietic activity was safely and transiently suppressed in adult sheep treated with 5 to 7.5 mg/kg BU. BU crossed the sheep placenta, and fetal sheep were indeed conditioned by administering 3 mg/kg BU to pregnant ewes. Engraftment of human CD34(+) cells in fetal recipients was enhanced when IUT was carried out 6 days post-BU. Up to 3.3% engraftment levels (in terms of bone marrow colony-forming units) were achieved with the IUT of 0.72 to 2.4 million CD34(+) cells when BU was used. BU can be administered to pregnant ewes to effectively condition the fetal recipient for IUT with enhanced engraftment of donor cells.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Bussulfano/administração & dosagem , Transplante de Células-Tronco Hematopoéticas , Modelos Animais , Animais , Antineoplásicos Fitogênicos/farmacocinética , Sequência de Bases , Bussulfano/farmacocinética , Primers do DNA , Feminino , Humanos , Troca Materno-Fetal , Placenta/metabolismo , Reação em Cadeia da Polimerase , Gravidez , Ovinos , Transplante Heterólogo
16.
Cell Med ; 3(1-3): 103-112, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28058187

RESUMO

Teratoma formation assays are established methods for evaluating the pluripotency of embryonic stem (ES) cells and induced pluripotent stem (iPS) cells. Teratoma formation in immunodeficient mice takes approximately 2 months. Here, we have developed a novel assay system for developing teratomas in vitro from ES cells and iPS cells in a short period. In vitro culture of ES, iPS, and mesenchymal stem cells (MSCs) in fetal rat metanephroi for 1 week resulted in distinct cell-dependent distribution patterns: Pluripotent cells (ES and iPS cells) formed aggregated masses, whereas MSCs showed disseminated distribution. The aggregated masses that had developed from ES cells and iPS cells after 2 weeks of culture comprised teratomas, though they were largely composed of immature components. Furthermore, in vitro organ culture for 1 week followed by relay transplantation into immunodeficient mice resulted in considerably rapid growing teratomas (teratomas developed in 4 weeks) having similar pathological features as of the teratomas developed using conventional 7-week in vivo teratoma formation assays. In addition, the initial cell number required in the in vitro assay was 1 × 103 cells, which was about 1% of the number of cells required in the conventional in vivo teratoma formation assays. These results suggest that the in vitro teratoma assay is a rapid and convenient screening system and might be an alternative method for developing teratomas for investigating the pluripotency of ES cells and iPS cells.

19.
Exp Hematol ; 39(1): 47-54, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20875838

RESUMO

OBJECTIVE: The homeobox B4 (HoxB4) gene promotes expansion of hematopoietic stem cells (HSCs). However, frequent development of leukemia in large animals due to retrovirally transduced HoxB4 gene has been reported. To prevent tumorigenesis, we developed a nonintegrating and nonreplicating Sendai virus vector that did not contain the phosphoprotein gene (SeV/ΔP), which enabled clearance of the vector and transgene shortly after transduction. We tested the SeV/ΔP vector expressing the HoxB4 gene (SeV/ΔP/HoxB4) for the ex vivo expansion of human cord blood CD34(+) cells (HSCs) using a sheep in utero transplantation assay. MATERIALS AND METHODS: Human HSCs were ex vivo-expanded by transduction with SeV/ΔP/HoxB4 vector and transplanted into the abdominal cavity of fetal sheep. The engraftment of human HSCs in the lambs was quantitatively evaluated by hematopoietic colony-forming unit assays. RESULTS: After transplantation, the HoxB4-transduced HSCs contributed to longer-period (up to 20 months) repopulation in sheep, and human hematopoietic progenitors were detected more frequently in the bone marrow of the HoxB4 group as compared with the control untreated group (p < 0.05). The expansion of human HSCs with the SeV/ΔP/HoxB4 vector was comparable with previously reported retroviral vectors expressing HoxB4. The SeV/ΔP/HoxB4 vector and the transgene were cleared from the recipient sheep and leukemia was not detected at 20 months post-transplantation. CONCLUSIONS: The SeV/ΔP vector would be suitable for transient expression of HoxB4 in human CD34(+) cells. In addition, the SeV/ΔP vector is free of concern about transgene-related and insertional leukemogenesis and should be safer than retroviral vectors.


Assuntos
Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Vírus Sendai/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Primers do DNA , Feminino , Humanos , Reação em Cadeia da Polimerase , Gravidez , Ovinos , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA