Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 12(1): 580, 2021 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-34802459

RESUMO

BACKGROUND: Redirection of natural killer (NK) cells with chimeric antigen receptors (CAR) is attractive in developing off-the-shelf CAR therapeutics for cancer treatment. However, the site-specific integration of a CAR gene into NK cells remains challenging. METHODS: In the present study, we genetically modified human induced pluripotent stem cells (iPSCs) with a zinc finger nuclease (ZFN) technology to introduce a cDNA encoding an anti-EpCAM CAR into the adeno-associated virus integration site 1, a "safe harbour" for transgene insertion into human genome, and next differentiated the modified iPSCs into CAR-expressing iNK cells. RESULTS: We detected the targeted integration in 4 out of 5 selected iPSC clones, 3 of which were biallelically modified. Southern blotting analysis revealed no random integration events. iNK cells were successfully derived from the modified iPSCs with a 47-day protocol, which were morphologically similar to peripheral blood NK cells, displayed NK phenotype (CD56+CD3-), and expressed NK receptors. The CAR expression of the iPSC-derived NK cells was confirmed with RT-PCR and flow cytometry analysis. In vitro cytotoxicity assay further confirmed their lytic activity against NK cell-resistant, EpCAM-positive cancer cells, but not to EpCAM-positive normal cells, demonstrating the retained tolerability of the CAR-iNK cells towards normal cells. CONCLUSION: Looking ahead, the modified iPSCs generated in the current study hold a great potential as a practically unlimited source to generate anti-EpCAM CAR iNK cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Receptores de Antígenos Quiméricos , Diferenciação Celular , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Matadoras Naturais , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo
2.
Mol Ther Oncolytics ; 17: 421-430, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32462079

RESUMO

Vγ9Vδ2 T cell-based anticancer immunotherapy has shown some promise in early-phase clinical trials but there is still large room for improvement. Using the extracellular domain of the human NKG2D, a stimulatory receptor expressed by Vγ9Vδ2 T cells, we constructed NKG2D ligand-specific chimeric antigen receptors (CARs). We adopted a non-viral CAR approach via mRNA electroporation to modify Vγ9Vδ2 T cells and demonstrated that, upon interaction with the NKG2D ligand-positive cancer cells, the CARs substantially enhanced the cytotoxic activity of the modified cells toward multiple cultured solid tumor cell lines, including those resistant to Zometa treatment. Repeated doses of the CAR-expressing cells resulted in tumor regression in mice with established tumors, extending median survival time by up to 132% as compared to the PBS control group. The findings suggest clinical potential for RNA CAR-modified Vγ9Vδ2 T cells to treat a wide variety of NKG2D ligand-expressing cancers.

3.
PLoS One ; 14(5): e0216815, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31071196

RESUMO

Using induced pluripotent stem cells (iPSCs) to derive chimeric antigen receptor-modified T (CAR-T) cells has great industrial potential. A previous study used αß T cell-derived CAR-modified iPSCs to produce CAR-T cells. However, these αß T cells are restricted to autologous use and only recognize single cancer antigen. To make CAR-T alternative for allogeneic use, we reprogrammed γδ T cell into iPSCs (γδ T-iPSCs) to circumvent the risk of graft-versus-host disease. To target multiple cancer-associated antigens, we used an "NK cell-promoting" protocol to differentiate γδ T-iPSCs and to induce expression of natural killer receptors (NKRs). Through such two-step strategy, mimetic γδ T cells endowed with an array of NKRs and thus designated as "γδ natural killer T (γδ NKT) cells" were derived. With no/low-level expression of inhibitory killer cell immunoglobulin-like receptors (KIRs) and immune checkpoint receptors, γδ NKT cells may provide a potent "off-the-shelf" cytotoxic cell source to recognize multiple ubiquitous antigens in a broad spectrum of cancers.


Assuntos
Antígenos de Neoplasias/imunologia , Células-Tronco Pluripotentes Induzidas/imunologia , Células T Matadoras Naturais/imunologia , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Antígenos Quiméricos/imunologia , Antígenos de Neoplasias/genética , Células HCT116 , Células Hep G2 , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células K562 , Células MCF-7 , Células T Matadoras Naturais/patologia , Neoplasias/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos Quiméricos/genética , Células THP-1
4.
Cytotherapy ; 20(3): 420-435, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29402645

RESUMO

Vγ9Vδ2 T cells are a minor subset of lymphocytes in the peripheral blood that has been extensively investigated for their tolerability, safety and anticancer efficacy. A hindrance to the broad application of these cells for adoptive cellular immunotherapy has been attaining clinically appropriate numbers of Vγ9Vδ2 T cells. Furthermore, Vγ9Vδ2 T cells exist at low frequencies among cancer patients. We, therefore, sought to conceive an economical method that allows for a quick and robust large-scale expansion of Vγ9Vδ2 T cells. A two-step protocol was developed, in which peripheral blood mononuclear cells (PBMCs) from healthy donors or cancer patients were activated with Zometa and interleukin (IL)-2, followed by co-culturing with gamma-irradiated, CD64-, CD86- and CD137L-expressing K562 artificial antigen-presenting cells (aAPCs) in the presence of the anti-CD3 antibody OKT3. We optimized the co-culture ratio of K562 aAPCs to immune cells, and migrated this method to a G-Rex cell growth platform to derive clinically relevant cell numbers in a Good Manufacturing Practice (GMP)-compliant manner. We further include a depletion step to selectively remove αß T lymphocytes. The method exhibited high expansion folds and a specific enrichment of Vγ9Vδ2 T cells. Expanded Vγ9Vδ2 T cells displayed an effector memory phenotype with a concomitant down-regulated expression of inhibitory immune checkpoint receptors. Finally, we ascertained the cytotoxic activity of these expanded cells by using nonmodified and chimeric antigen receptor (CAR)-engrafted Vγ9Vδ2 T cells against a panel of solid tumor cells. Overall, we report an efficient approach to generate highly functional Vγ9Vδ2 T cells in massive numbers suitable for clinical application in an allogeneic setting.


Assuntos
Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/citologia , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/metabolismo , Complexo CD3/imunologia , Proliferação de Células/efeitos dos fármacos , Células Alimentadoras/metabolismo , Humanos , Imunofenotipagem , Interleucina-2/farmacologia , Células K562 , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
Stem Cell Reports ; 9(6): 1796-1812, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29173894

RESUMO

Current donor cell-dependent strategies can only produce limited "made-to-order" therapeutic natural killer (NK) cells for limited patients. To provide unlimited "off-the-shelf" NK cells that serve many recipients, we designed and demonstrated a holistic manufacturing scheme to mass-produce NK cells from induced pluripotent stem cells (iPSCs). Starting with a highly accessible human cell source, peripheral blood cells (PBCs), we derived a good manufacturing practice-compatible iPSC source, PBC-derived iPSCs (PBC-iPSCs) for this purpose. Through our original protocol that excludes CD34+ cell enrichment and spin embryoid body formation, high-purity functional and expandable NK cells were generated from PBC-iPSCs. Above all, most of these NK cells expressed no killer cell immunoglobulin-like receptors (KIRs), which renders them unrestricted by recipients' human leukocyte antigen genotypes. Hence, we have established a practical "from blood cell to stem cells and back with less (less KIRs)" strategy to generate abundant "universal" NK cells from PBC-iPSCs for a wide range of patients.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células Matadoras Naturais/citologia , Proliferação de Células/genética , Corpos Embrioides/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Matadoras Naturais/metabolismo , Células-Tronco de Sangue Periférico/citologia , Células-Tronco de Sangue Periférico/metabolismo , Receptores KIR/genética
6.
Sci Rep ; 5: 15262, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26471005

RESUMO

Human pluripotent stem cells (hPSCs) provide a promising platform to produce dendritic cell (DC) vaccine. To streamline the production process, we investigated a unique antigen-loading strategy that suits this novel platform. Specifically, we stably modified hPSCs using tumour antigen genes in the form of a full-length tumour antigen gene or an artificial tumour antigen epitope-coding minigene. Such antigenically modified hPSCs were able to differentiate into tumour antigen-presenting DCs. Without conventional antigen-loading, DCs derived from the minigene-modified hPSCs were ready to prime a tumour antigen-specific T cell response and further expand these specific T cells in restimulation processes. These expanded tumour antigen-specific T cells were potent effectors with central memory or effector memory phenotype. Thus, we demonstrated that immunocompetent tumour antigen-loaded DCs can be directly generated from antigenically modified hPSCs. Using such strategy, we can completely eliminate the conventional antigen-loading step and significantly simplify the production of DC vaccine from hPSCs.


Assuntos
Antígenos de Neoplasias/metabolismo , Células Dendríticas/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/imunologia , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Epitopos/genética , Epitopos/imunologia , Epitopos/metabolismo , Genes Reporter , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Imunocompetência , Antígeno MART-1/genética , Antígeno MART-1/imunologia , Antígeno MART-1/metabolismo , Neoplasias/terapia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Elementos de Resposta/genética
7.
Stem Cells Transl Med ; 3(8): 923-35, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24972599

RESUMO

Given their intrinsic ability to home to tumor sites, endothelial progenitor cells (EPCs) are attractive as cellular vehicles for targeted cancer gene therapy. However, collecting sufficient EPCs is one of the challenging issues critical for effective clinical translation of this new approach. In this study, we sought to explore whether human induced pluripotent stem (iPS) cells could be used as a reliable and accessible cell source to generate human EPCs suitable for cancer treatment. We used an embryoid body formation method to derive CD133(+)CD34(+) EPCs from human iPS cells. The generated EPCs expressed endothelial markers such as CD31, Flk1, and vascular endothelial-cadherin without expression of the CD45 hematopoietic marker. After intravenous injection, the iPS cell-derived EPCs migrated toward orthotopic and lung metastatic tumors in the mouse 4T1 breast cancer model but did not promote tumor growth and metastasis. To investigate their therapeutic potential, the EPCs were transduced with baculovirus encoding the potent T cell costimulatory molecule CD40 ligand. The systemic injection of the CD40 ligand-expressing EPCs stimulated the secretion of both tumor necrosis factor-α and interferon-γ and increased the caspase 3/7 activity in the lungs with metastatic tumors, leading to prolonged survival of the tumor bearing mice. Therefore, our findings suggest that human iPS cell-derived EPCs have the potential to serve as tumor-targeted cellular vehicles for anticancer gene therapy.


Assuntos
Neoplasias da Mama/terapia , Ligante de CD40/biossíntese , Células Endoteliais/transplante , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/transplante , Neoplasias Pulmonares/terapia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Baculoviridae/genética , Baculoviridae/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ligante de CD40/genética , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Células Endoteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Camundongos Nus , Peptídeos/metabolismo , Fatores de Tempo , Transdução Genética , Transfecção , Carga Tumoral
8.
Hum Gene Ther ; 25(8): 747-58, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24773154

RESUMO

The interaction between CD40 ligand (CD40L) and CD40 can directly inhibit growth of CD40-positive carcinoma cells and may indirectly inhibit tumor growth through coordination of immune responses. Many efforts in CD40L cancer gene therapy have been focused on direct CD40L gene transfer into malignant target cells. This in vivo gene therapy approach relies on high-efficiency gene transfer and could be technically challenging for the treatment of certain cancers, especially multisite metastases. We report herein an alternative means of using the tumor-homing property of neural stem cells (NSCs) to deliver CD40L molecules into tumor tissues. NSCs were derived from human induced pluripotent stem cells, transduced in vitro with a baculoviral vector encoding CD40L, and intravenously injected into immunocompetent mice with orthotopic and metastatic breast cancers. Through a bystander mechanism of intercellular transfer of CD40L from the donor NSCs to tumor target cells, the treatment impeded tumor growth, leading to prolonged survival of the tumor-bearing mice. We further showed that compared with the stem cell-based gene therapy that employed a suicide gene, the CD40L immunogene therapy did not cause liver and kidney injury in the treated mice. This new approach may be particularly valuable for metastatic cancer treatments after systemic stem cell administration.


Assuntos
Baculoviridae/genética , Neoplasias da Mama/terapia , Ligante de CD40/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Neoplasias Pulmonares/terapia , Células-Tronco Neurais/metabolismo , Animais , Apoptose , Neoplasias da Mama/patologia , Ligante de CD40/biossíntese , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Transdução Genética
9.
Stem Cells Transl Med ; 3(1): 69-80, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24292792

RESUMO

Invariant natural killer T (iNKT) cells are a unique lymphocyte subpopulation that mediates antitumor activities upon activation. A current strategy to harness iNKT cells for cancer treatment is endogenous iNKT cell activation using patient-derived dendritic cells (DCs). However, the limited number and functional defects of patient DCs are still the major challenges for this therapeutic approach. In this study, we investigated whether human embryonic stem cells (hESCs) with an ectopically expressed CD1d gene could be exploited to address this issue. Using a lentivector carrying an optimized expression cassette, we generated stably modified hESC lines that consistently overexpressed CD1d. These modified hESC lines were able to differentiate into DCs as efficiently as the parental line. Most importantly, more than 50% of such derived DCs were CD1d+. These CD1d-overexpressing DCs were more efficient in inducing iNKT cell response than those without modification, and their ability was comparable to that of DCs generated from monocytes of healthy donors. The iNKT cells expanded by the CD1d-overexpressing DCs were functional, as demonstrated by their ability to lyse iNKT cell-sensitive glioma cells. Therefore, hESCs stably modified with the CD1d gene may serve as a convenient, unlimited, and competent DC source for iNKT cell-based cancer immunotherapy.


Assuntos
Antígenos CD1d/genética , Neoplasias Encefálicas/terapia , Células Dendríticas/citologia , Glioma/terapia , Imunoterapia/métodos , Células T Matadoras Naturais/citologia , Antígenos CD1d/metabolismo , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Divisão Celular/imunologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Citometria de Fluxo , Expressão Gênica/imunologia , Glioma/imunologia , Glioma/patologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Células T Matadoras Naturais/imunologia , Transgenes/imunologia
10.
Stem Cells Transl Med ; 2(12): 935-45, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24167318

RESUMO

Integrative gene transfer using retroviruses to express reprogramming factors displays high efficiency in generating induced pluripotent stem cells (iPSCs), but the value of the method is limited because of the concern over mutagenesis associated with random insertion of transgenes. Site-specific integration into a preselected locus by engineered zinc-finger nuclease (ZFN) technology provides a potential way to overcome the problem. Here, we report the successful reprogramming of human fibroblasts into a state of pluripotency by baculoviral transduction-mediated, site-specific integration of OKSM (Oct3/4, Klf4, Sox2, and c-myc) transcription factor genes into the AAVS1 locus in human chromosome 19. Two nonintegrative baculoviral vectors were used for cotransduction, one expressing ZFNs and another as a donor vector encoding the four transcription factors. iPSC colonies were obtained at a high efficiency of 12% (the mean value of eight individual experiments). All characterized iPSC clones carried the transgenic cassette only at the ZFN-specified AAVS1 locus. We further demonstrated that when the donor cassette was flanked by heterospecific loxP sequences, the reprogramming genes in iPSCs could be replaced by another transgene using a baculoviral vector-based Cre recombinase-mediated cassette exchange system, thereby producing iPSCs free of exogenous reprogramming factors. Although the use of nonintegrating methods to generate iPSCs is rapidly becoming a standard approach, methods based on site-specific integration of reprogramming factor genes as reported here hold the potential for efficient generation of genetically amenable iPSCs suitable for future gene therapy applications.


Assuntos
Baculoviridae/genética , Reprogramação Celular , Endonucleases/metabolismo , Fibroblastos/metabolismo , Vetores Genéticos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fatores de Transcrição/metabolismo , Transdução Genética , Dedos de Zinco , Diferenciação Celular , Linhagem Celular , Cromossomos Humanos Par 19 , Endonucleases/genética , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Integrases/genética , Integrases/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/genética , Transfecção
11.
Mol Ther ; 21(8): 1621-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23752308

RESUMO

Intravenously injected neural stem cells (NSCs) can infiltrate both primary and metastatic tumor sites; thus, they are attractive tumor-targeting vehicles for delivering anticancer agents. However, because the systemic distribution of the injected NSCs involves normal organs and might induce off-target actions leading to unintended side effects, clinical applications of this approach is impeded. Given that the vesicular stomatitis virus glycoprotein (VSV-G) can promote the formation of multinucleated syncytia to kill cells in a pH-dependent manner, we engineered a pH sensor of VSV-G and generated a novel VSV-G mutant that efficiently promotes syncytium formation at the tumor extracellular pH (pHe) but not at pH 7.4. Using transduced NSCs derived from induced pluripotent stem cells (iPSCs), the VSV-G mutant was delivered into mice with metastatic breast cancers in the lung through tail vein injection. Compared with the conventional stem cell-based gene therapy that uses the herpes simplex virus thymidine kinase (HSVtk) suicide gene, this treatment did not display toxicity to normal non-targeted organs while retaining therapeutic effects in tumor-bearing organs. Our findings demonstrate the effectiveness of a new approach for achieving tumor-selective killing effects following systemic stem cell administration. Its potential in stem cell-based gene therapy for metastatic cancer is worthy of further exploration.


Assuntos
Glicoproteínas de Membrana/genética , Neoplasias/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Proteínas do Envelope Viral/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Morte Celular , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Feminino , Genes Transgênicos Suicidas , Terapia Genética , Células Gigantes/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Células-Tronco Pluripotentes Induzidas/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Metástase Neoplásica , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Proteínas do Envelope Viral/metabolismo
12.
J Immunol ; 188(9): 4297-304, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22407918

RESUMO

Human embryonic stem cell-derived dendritic cells (hESC-DCs) may potentially provide a platform to generate "off-the-shelf" therapeutic cancer vaccines. To apply hESC-DCs for cancer immunotherapy in a semiallogeneic setting, it is crucial for these cells to "jump-start" adaptive antitumor immunity before their elimination by host alloreaction. In this study, we investigated whether CD1d upregulation in hESC-DCs may exploit invariant NKT (iNKT) cell adjuvant activity and boost antitumor immunity. Using a baculoviral vector carrying the CD1d gene, we produced CD1d-overexpressing hESC-DCs and demonstrated that the upregulated CD1d was functional in presenting α-galactosylceramide for iNKT cell expansion. Pulsed with melanoma Ag recognized by T cell 1 peptide, the CD1d-overexpressing hESC-DCs displayed enhanced capability to prime CD8(+) T cells without relying on α-galactosylceramide loading. Blocking the CD1d with Ab reduced the immunogenicity, suggesting the importance of hESC-DC and iNKT cell interaction in this context. The CD1d-overexpressing hESC-DCs also induced a proinflammatory cytokine profile that may favor the T cell priming. Moreover, a similar immunostimulatory effect was observed when the CD1d upregulation strategy was applied in human monocyte-derived dendritic cells. Therefore, our study suggests that the upregulation of CD1d in hESC-DCs provides a novel strategy to enhance their immunogenicity. This approach holds potential for advancing the application of hESC-DCs into human cancer immunotherapy.


Assuntos
Antígenos CD1d/imunologia , Comunicação Celular/imunologia , Células Dendríticas/imunologia , Células-Tronco Embrionárias/imunologia , Regulação para Cima/imunologia , Animais , Antígenos CD1d/biossíntese , Antígenos CD1d/genética , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/metabolismo , Comunicação Celular/genética , Linhagem Celular Tumoral , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Regulação para Cima/genética
13.
J Biosci Bioeng ; 109(1): 1-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20129073

RESUMO

Transient gene expression is one possible approach to manipulate the signaling pathways that control the proliferation and differentiation of human embryonic stem (hES) cells. We tested in hES cells a range of baculoviral vectors with a human elongation factor-1alpha promoter and various viral regulatory elements and observed the most dramatic augmenting effect on the transient expression when the promoter was used together with the human cytomegalovirus immediate-early gene enhancer and the woodchuck hepatitis virus post-transcriptional regulatory elements. This vector provided a 1.6-fold increase in the percentage of transduced cells (up to 72%) over a vector containing the elongation factor-1alpha promoter alone. The effective baculoviral transduction of hES cells did not affect cell proliferation, expression of embryonic stem cell markers and teratoma formation. This new viral vector for temporary transgene expression might become a useful tool for developmental biology studies and biomedical applications of hES cells.


Assuntos
Baculoviridae/genética , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos/genética , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas/genética , Transdução Genética/métodos , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Células Cultivadas , Citomegalovirus/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/virologia , Genes Precoces/genética , Vírus da Hepatite B da Marmota/genética , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Fator 1 de Elongação de Peptídeos/metabolismo , Teratoma/metabolismo , Transgenes/genética
14.
Mol Ther ; 17(12): 2058-66, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19809402

RESUMO

Transcriptional targeting using a tissue-specific cellular promoter is proving to be a powerful means for restricting transgene expression in targeted tissues. In the context of cancer suicide gene therapy, this approach may lead to cytotoxic effects in both cancer and nontarget normal cells. Considering microRNA (miRNA) function in post-transcriptional regulation of gene expression, we have developed a viral vector platform combining cellular promoter-based transcriptional targeting with miRNA regulation for a glioma suicide gene therapy in the mouse brain. The therapy employed, in a single baculoviral vector, a glial fibrillary acidic protein (GFAP) gene promoter and the repeated target sequences of three miRNAs that are enriched in astrocytes but downregulated in glioblastoma cells to control the expression of the herpes simplex virus thymidine kinase (HSVtk) gene. This resulted in significantly improved in vivo selectivity over the use of a control vector without miRNA regulation, enabling effective elimination of human glioma xenografts while producing negligible toxic effects on normal astrocytes. Thus, incorporating miRNA regulation into a transcriptional targeting vector adds an extra layer of security to prevent off-target transgene expression and should be useful for the development of gene delivery vectors with high targeting specificity for cancer therapy.


Assuntos
Neoplasias Encefálicas/terapia , Regulação Neoplásica da Expressão Gênica , Genes Transgênicos Suicidas/fisiologia , Terapia Genética , Glioblastoma/terapia , MicroRNAs/fisiologia , Regiões Promotoras Genéticas/genética , Animais , Astrócitos/metabolismo , Baculoviridae/genética , Neoplasias Encefálicas/genética , Células Cultivadas , Quimioterapia Combinada , Feminino , Vetores Genéticos , Proteína Glial Fibrilar Ácida/genética , Glioblastoma/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Timidina Quinase/genética , Transgenes/fisiologia
15.
Mol Ther ; 17(9): 1585-93, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19532141

RESUMO

Transient genetic manipulation of human neurons without chromosomal integration of the transgene would be valuable but has been challenging due to the quiescent nature of these postmitotic cells. In this study, we developed a set of baculoviral vectors for transient transduction in nondividing neurons derived from human embryonic stem cells (hESCs). Using a baculoviral vector equipped with the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), we observed a quick onset of transgene expression as early as day 1 after baculoviral transduction and a high efficiency of up to 80%. Strong transgene expression in the cultured human neurons was observed for more than 1 month and the signal was easily detectable even after 3 months. Using two baculoviral vectors carrying different transgenes, we found that co-transduction at a single neuron level was possible. After transplantation into the brain of nude mice, the baculovirus-transduced human neurons were integrated into the mouse brain and maintained transgene expression for at least 4 weeks, portending the usefulness of this technique in assisting neural transplantation. Therefore, by mediating efficient transient gene expression, baculoviral vectors can provide useful tools for both basic gene function studies in human neurons and therapeutic applications of these cells.


Assuntos
Baculoviridae/genética , Células-Tronco Embrionárias/citologia , Neurônios/metabolismo , Transdução Genética/métodos , Animais , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Genes Virais/genética , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Vírus da Hepatite B da Marmota/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Neurônios/citologia
16.
Mol Ther ; 17(6): 1003-11, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19240692

RESUMO

Achievement of specific tumor cell targeting remains a challenge for glioma gene therapy. We observed that the human high mobility group box2 (HMGB2) gene had a low level of expression in normal human brain tissues, but was significantly upregulated in glioblastoma tissues. With progressive truncation of a 5'-upstream sequence of the HMGB2 gene, we identified a 0.5-kb fragment displaying a high transcriptional activity in glioblastoma cells, but a low activity in normal brain cells. To test the feasibility of using the HMGB2 promoter sequence in targeted cancer therapy, we constructed a baculoviral vector expressing the herpes simplex virus thymidine kinase (HSVtk) gene driven by the HMGB2 promoter. Transduction with the viral vector induced cell death in glioblastoma cell lines in the presence of ganciclovir (GCV), but did not affect the survival of human astrocytes and neurons. In a mouse xenograft model, intratumor injection of the baculoviral vector suppressed the growth of human glioblastoma cells and prolonged the survival of tumor-bearing mice. Our results suggest that the novel 5' sequence of HMGB2 gene has a potential to be used as an efficient, tumor-selective promoter in targeted vectors for glioblastoma gene therapy.


Assuntos
Genes Transgênicos Suicidas/fisiologia , Terapia Genética/métodos , Glioblastoma/terapia , Proteína HMGB2/genética , Regiões Promotoras Genéticas/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Ganciclovir/uso terapêutico , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Transgênicos Suicidas/genética , Vetores Genéticos/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/fisiologia , Transcrição Gênica/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Stem Cells ; 25(4): 1055-61, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17420229

RESUMO

Human embryonic stem (hES) cells as a renewable cell source have great prospective applications in both developmental biology research and regenerative medicine. To realize these potentials, the development of effective and safe genetic manipulation methods in hES cells is an obvious demand. We report here that baculoviral vectors were able to transduce hES cells efficiently. In transient transduction experiments, a recombinant baculoviral vector equipped with a human elongation factor 1-alpha promoter and a woodchuck hepatitis post-transcriptional regulatory element transduced up to 80% of cells in hES cell clumps and embryoid bodies. For prolonged transgene expression, hybrid baculoviral vectors that have incorporated a rep gene and inverted terminal repeat sequences from adeno-associated virus were produced. These hybrid vectors yielded stable transgene expression during the prolonged undifferentiated proliferation of hES cells and after differentiation. Baculoviral transduction did not affect the normal growth, phenotype, and pluripotency of hES cells. Thus, baculoviral vectors suitable for both transient overexpression and long-term stable expression are an attractive option for genetic manipulation of hES cells.


Assuntos
Baculoviridae/genética , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/transplante , Vetores Genéticos , Células-Tronco Embrionárias/virologia , Citometria de Fluxo , Regulação da Expressão Gênica , Genes Reporter , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imuno-Histoquímica , Proteínas Recombinantes de Fusão , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Transfecção , Transgenes
18.
Biomaterials ; 28(7): 1443-51, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17156837

RESUMO

Chemical conjugation of targeting ligands to polycation/plasmid DNA complexes has been widely used to improve the transfection efficiency of nonviral gene delivery vectors. However, conjugation reactions may reduce or even inactivate the biological activities of chemically sensitive moieties, such as proteins and peptides. Here we describe a new method for introducing targeting ligands into nonviral vectors, in which ternary complexes are formed via charge interactions among polyethylenimine (PEI) of 600Da, plasmid DNA and targeting peptides with positively charged DNA-binding sequence. Owing to the nerve growth factor (NGF) loop 4 hairpin motif in the targeting peptide, these ternary complexes are capable of mediating gene delivery efficiently and specifically into cells expressing the NGF receptor TrkA. In in vitro experiments, the complexes improved luciferase reporter gene expression by up to 1000-fold while comparing with that produced by complexes with nontargeting control peptide. In an in vivo experiment, the ternary complexes with the targeting peptide was 59-fold more efficient than the control ternary complexes in transfecting dorsal root ganglia (DRG), the peripheral nervous sites with TrkA-expressing neurons. In a cell viability study, the ternary complexes were remarkably different from DNA complexes by PEI of 25 kDa, the gold standard for nonviral gene carriers, displaying no toxicity in tested neuronal cells. Thus, this study demonstrates an alternative method to construct nonviral delivery system for targeted gene transfer into neurons.


Assuntos
Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Peptídeos/administração & dosagem , Plasmídeos/administração & dosagem , Animais , Materiais Biocompatíveis/química , Células Cultivadas , Sistemas de Liberação de Medicamentos , Genes Reporter , Terapia Genética , Luciferases/genética , Teste de Materiais , Camundongos , Peso Molecular , Células NIH 3T3 , Plasmídeos/genética , Polietilenoimina/química , Ratos , Receptor trkA/metabolismo
19.
CSH Protoc ; 2006(1)2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22485782

RESUMO

INTRODUCTION This protocol describes the preparation of polyethylenimine (PEI)/DNA nanoparticles for targeted gene delivery. This delivery strategy improves the efficiency of gene transfer by enhancing the entry of gene vectors into the desired cells and reducing uptake by nontarget cells. We describe here methods for the conjugation of targeting peptides to PEIs, formation of DNA complexes using the conjugated PEIs or nonconjugated PEIs together with targeting peptides, and cell transfection using these complexes. The conjugation step involves the use of the succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), a heterobifunctional cross-linker, to form a stable bond between PEI and peptides containing thiol groups.

20.
Mol Ther ; 12(2): 314-20, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15925545

RESUMO

Gene delivery to sensory neurons of the dorsal root ganglion (DRG) offers the prospect of developing new clinical interventions against peripheral nerve diseases and disorders. Here we show that genes can be transferred to rat DRG through lumbar intrathecal injection of delivery vectors into the cerebrospinal fluid. Genes could be transferred to DRG using polyethylenimine (PEI)/DNA complexes, Lipofectamine 2000/DNA complexes, adeno-associated virus vectors, or baculovirus vectors. We also show that nerve growth factor cDNA, delivered through lumbar intrathecal injection of PEI complexes, was able to improve regeneration of transected rat sciatic nerves. These data demonstrate the viability of using an intrathecal gene delivery approach for treating peripheral neuropathies.


Assuntos
DNA Complementar/metabolismo , Gânglios Espinais/fisiologia , Técnicas de Transferência de Genes , Regeneração Nervosa , Nervo Isquiático/fisiologia , Animais , Baculoviridae/genética , Dependovirus/genética , Gânglios Espinais/metabolismo , Expressão Gênica , Genes Reporter , Vetores Genéticos , Injeções Espinhais , Lipídeos/genética , Lipossomos , Luciferases/genética , Luciferases/metabolismo , Masculino , Fatores de Crescimento Neural , Regeneração Nervosa/genética , Polietilenoimina , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...