Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Vis Exp ; (117)2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27911363

RESUMEN

Adoptive transfer of ex vivo expanded autologous tumor-infiltrating lymphocytes (TILs) can mediate durable and complete responses in significant subsets of patients with metastatic melanoma. Major obstacles of this approach are the reduced viability of transferred T cells, caused by telomere shortening, and the limited number of TILs obtained from patients. Less-differentiated T cells with long telomeres would be an ideal T cell subset for adoptive T cell therapy;however, generating large numbers of these less-differentiated T cells is problematic. This limitation of adoptive T cell therapy can be theoretically overcome by using induced pluripotent stem cells (iPSCs) that self-renew, maintain pluripotency, have elongated telomeres, and provide an unlimited source of autologous T cells for immunotherapy. Here, we present a protocol to generate iPSCs using Sendai virus vectors for the transduction of reprogramming factors into TILs. This protocol generates fully reprogrammed, vector-free clones. These TIL-derived iPSCs might be able to generate less-differentiated patient- and tumor-specific T cells for adoptive T cell therapy.


Asunto(s)
Células Madre Pluripotentes Inducidas , Linfocitos Infiltrantes de Tumor , Melanoma/patología , Traslado Adoptivo , Humanos , Inmunoterapia Adoptiva
2.
Stem Cells Int ; 2016: 8394960, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057178

RESUMEN

Induced pluripotent stem cells (iPSCs) derived from somatic cells of patients hold great promise for autologous cell therapies. One of the possible applications of iPSCs is to use them as a cell source for producing autologous lymphocytes for cell-based therapy against cancer. Tumor-infiltrating lymphocytes (TILs) that express programmed cell death protein-1 (PD-1) are tumor-reactive T cells, and adoptive cell therapy with autologous TILs has been found to achieve durable complete response in selected patients with metastatic melanoma. Here, we describe the derivation of human iPSCs from melanoma TILs expressing high level of PD-1 by Sendai virus-mediated transduction of the four transcription factors, OCT3/4, SOX2, KLF4, and c-MYC. TIL-derived iPSCs display embryonic stem cell-like morphology, have normal karyotype, express stem cell-specific surface antigens and pluripotency-associated transcription factors, and have the capacity to differentiate in vitro and in vivo. A wide variety of T cell receptor gene rearrangement patterns in TIL-derived iPSCs confirmed the heterogeneity of T cells infiltrating melanomas. The ability to reprogram TILs containing patient-specific tumor-reactive repertoire might allow the generation of patient- and tumor-specific polyclonal T cells for cancer immunotherapy.

3.
Stem Cells Transl Med ; 5(5): 694-702, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26987352

RESUMEN

UNLABELLED: Human induced pluripotent stem cells (iPSCs) and derived progeny provide invaluable regenerative platforms, yet their clinical translation has been compromised by their biosafety concern. Here, we assessed the safety of transplanting patient-derived iPSC-generated pancreatic endoderm/progenitor cells. Transplantation of progenitors from iPSCs reprogrammed by lentiviral vectors (LV-iPSCs) led to the formation of invasive teratocarcinoma-like tumors in more than 90% of immunodeficient mice. Moreover, removal of primary tumors from LV-iPSC progeny-transplanted hosts generated secondary and metastatic tumors. Combined transgene-free (TGF) reprogramming and elimination of residual pluripotent cells by enzymatic dissociation ensured tumor-free transplantation, ultimately enabling regeneration of type 1 diabetes-specific human islet structures in vivo. The incidence of tumor formation in TGF-iPSCs was titratable, depending on the oncogenic load, with reintegration of the cMYC expressing vector abolishing tumor-free transplantation. Thus, transgene-free cMYC-independent reprogramming and elimination of residual pluripotent cells are mandatory steps in achieving transplantation of iPSC progeny for customized and safe islet regeneration in vivo. SIGNIFICANCE: Pluripotent stem cell therapy for diabetes relies on the safety as well as the quality of derived insulin-producing cells. Data from this study highlight prominent tumorigenic risks of induced pluripotent stem cell (iPSC) products, especially when reprogrammed with integrating vectors. Two major underlying mechanisms in iPSC tumorigenicity are residual pluripotent cells and cMYC overload by vector integration. This study also demonstrated that combined transgene-free reprogramming and enzymatic dissociation allows teratoma-free transplantation of iPSC progeny in the mouse model in testing the tumorigenicity of iPSC products. Further safety assessment and improvement in iPSC specification into a mature ß cell phenotype would lead to safe islet replacement therapy for diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Diabetes Mellitus Tipo 2/cirugía , Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/cirugía , Queratinocitos/trasplante , Regeneración , Teratocarcinoma/prevención & control , Adulto , Anciano , Animales , Diferenciación Celular , Células Cultivadas , Técnicas de Reprogramación Celular , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Xenoinjertos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos/efectos adversos , Queratinocitos/metabolismo , Queratinocitos/patología , Lentivirus/genética , Masculino , Ratones SCID , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Teratocarcinoma/genética , Teratocarcinoma/metabolismo , Teratocarcinoma/patología , Transfección
4.
Cell Mol Life Sci ; 73(10): 2089-104, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26573968

RESUMEN

Spinal muscular atrophy (SMA) is a devastating motoneuron (MN) disorder caused by homozygous loss of SMN1. Rarely, SMN1-deleted individuals are fully asymptomatic despite carrying identical SMN2 copies as their SMA III-affected siblings suggesting protection by genetic modifiers other than SMN2. High plastin 3 (PLS3) expression has previously been found in lymphoblastoid cells but not in fibroblasts of asymptomatic compared to symptomatic siblings. To find out whether PLS3 is also upregulated in MNs of asymptomatic individuals and thus a convincing SMA protective modifier, we generated induced pluripotent stem cells (iPSCs) from fibroblasts of three asymptomatic and three SMA III-affected siblings from two families and compared these to iPSCs from a SMA I patient and control individuals. MNs were differentiated from iPSC-derived small molecule neural precursor cells (smNPCs). All four genotype classes showed similar capacity to differentiate into MNs at day 8. However, SMA I-derived MN survival was significantly decreased while SMA III- and asymptomatic-derived MN survival was moderately reduced compared to controls at day 27. SMN expression levels and concomitant gem numbers broadly matched SMN2 copy number distribution; SMA I presented the lowest levels, whereas SMA III and asymptomatic showed similar levels. In contrast, PLS3 was significantly upregulated in mixed MN cultures from asymptomatic individuals pinpointing a tissue-specific regulation. Evidence for strong PLS3 accumulation in shaft and rim of growth cones in MN cultures from asymptomatic individuals implies an important role in neuromuscular synapse formation and maintenance. These findings provide strong evidence that PLS3 is a genuine SMA protective modifier.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/genética , Células-Madre Neurales/citología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Regulación hacia Arriba , Biopsia , Diferenciación Celular , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Silenciador del Gen , Vectores Genéticos , Genotipo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Cariotipificación , Linfocitos/citología , Masculino , Microscopía Confocal , Mutación , Linaje , Fenotipo , ARN Interferente Pequeño/metabolismo , Piel/patología
5.
Stem Cells ; 33(6): 2077-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25694335

RESUMEN

Trisomy 21 (T21), Down Syndrome (DS) is the most common genetic cause of dementia and intellectual disability. Modeling DS is beginning to yield pharmaceutical therapeutic interventions for amelioration of intellectual disability, which are currently being tested in clinical trials. DS is also a unique genetic system for investigation of pathological and protective mechanisms for accelerated ageing, neurodegeneration, dementia, cancer, and other important common diseases. New drugs could be identified and disease mechanisms better understood by establishment of well-controlled cell model systems. We have developed a first nonintegration-reprogrammed isogenic human induced pluripotent stem cell (iPSC) model of DS by reprogramming the skin fibroblasts from an adult individual with constitutional mosaicism for DS and separately cloning multiple isogenic T21 and euploid (D21) iPSC lines. Our model shows a very low number of reprogramming rearrangements as assessed by a high-resolution whole genome CGH-array hybridization, and it reproduces several cellular pathologies seen in primary human DS cells, as assessed by automated high-content microscopic analysis. Early differentiation shows an imbalance of the lineage-specific stem/progenitor cell compartments: T21 causes slower proliferation of neural and faster expansion of hematopoietic lineage. T21 iPSC-derived neurons show increased production of amyloid peptide-containing material, a decrease in mitochondrial membrane potential, and an increased number and abnormal appearance of mitochondria. Finally, T21-derived neurons show significantly higher number of DNA double-strand breaks than isogenic D21 controls. Our fully isogenic system therefore opens possibilities for modeling mechanisms of developmental, accelerated ageing, and neurodegenerative pathologies caused by T21.


Asunto(s)
Envejecimiento/fisiología , Diferenciación Celular/fisiología , Síndrome de Down/genética , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Animales , Células Cultivadas , Fibroblastos/citología , Humanos , Mitocondrias/genética
6.
Stem Cell Res ; 12(2): 574-83, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24531302

RESUMEN

Familial amyloidotic polyneuropathy (FAP) is a hereditary amyloidosis induced by amyloidogenic transthyretin (ATTR). Because most transthyretin (TTR) in serum is synthesized by the liver, liver transplantation (LT) is today the only treatment available to halt the progression of FAP, even though LT is associated with several problems. Despite the urgent need to develop alternatives to LT, the detailed pathogenesis of FAP is still unknown; also, no model fully represents the relevant processes in patients with FAP. The induction of induced pluripotent stem (iPS) cells has allowed development of pluripotent cells specific for patients and has led to useful models of human diseases. Because of the need for a tool to elucidate the molecular pathogenesis of FAP, in this study we sought to establish heterozygous ATTR mutant iPS cells, and were successful, by using a Sendai virus vector mixture containing four transcription factors (Oct3/4, Sox2, Klf4, and c-Myc) to reprogram dermal fibroblasts derived from FAP patients. Moreover, FAP-specific iPS cells had the potential to differentiate into hepatocyte-like cells and indeed expressed ATTR. FAP-specific iPS cells demonstrated the possibility of serving as a pathological tool that will contribute to understanding the pathogenesis of FAP and development of FAP treatments.


Asunto(s)
Neuropatías Amiloides Familiares/patología , Células Madre Pluripotentes Inducidas/patología , Prealbúmina/metabolismo , Neuropatías Amiloides Familiares/genética , Animales , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones
7.
Cell Reprogram ; 15(6): 503-13, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24219578

RESUMEN

Studies of rare genetic bone disorders are often limited due to unavailability of tissue specimens and the lack of animal models fully replicating phenotypic features. Craniometaphyseal dysplasia (CMD) is a rare monogenic disorder characterized by hyperostosis of craniofacial bones concurrent with abnormal shape of long bones. Mutations for autosomal dominant CMD have been identified in the ANK gene (ANKH). Here we describe a simple and efficient method to reprogram adherent cells cultured from peripheral blood to human induced pluripotent stem cells (hiPSCs) from eight CMD patients and five healthy controls. Peripheral blood mononuclear cells (PBMCs) were separated from 5-7 mL of whole blood by Ficoll gradient, expanded in the presence of cytokines and transduced with Sendai virus (SeV) vectors encoding OCT3/4, SOX2, KLF4, and c-MYC. SeV vector, a cytoplasmic RNA vector, is lost from host cells after propagation for 10-13 passages. These hiPSCs express stem cell markers, have normal karyotypes, and are capable of forming embryoid bodies in vitro as well as teratomas in vivo. Further differentiation of these patient-specific iPSCs into osteoblasts and osteoclasts can provide a useful tool to study the effects CMD mutations on bone, and this approach can be applied for disease modeling of other rare genetic musculoskeletal disorders.


Asunto(s)
Enfermedades del Desarrollo Óseo/sangre , Reprogramación Celular , Anomalías Craneofaciales/sangre , Vectores Genéticos , Hiperostosis/sangre , Hipertelorismo/sangre , Células Madre Pluripotentes Inducidas/citología , Virus Sendai/genética , Adulto , Secuencia de Bases , Estudios de Casos y Controles , Niño , Cartilla de ADN , Femenino , Reordenamiento Génico de Linfocito T , Humanos , Factor 4 Similar a Kruppel , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
8.
Oncol Lett ; 6(2): 323-328, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24137324

RESUMEN

Cancer is initiated by the transformation of stem cells or progenitor cells via a dedifferentiation process that leads to cancer stem cells; however, the process involves the activation of growth-promoting oncogenes and the inactivation of growth-constraining tumor suppressor genes. The introduction of defined factors, such as those encoded by c-Myc, Sox2, Oct3/4 and Klf4, in normal somatic cells results in their dedifferentiation into induced pluripotent stem (iPS) cells. We previously reported that these defined factors induced the development of induced multipotent cancer (iPC) cells from gastrointestinal cancer cells by reducing tumor aggressiveness. Previous studies indicated that although reprogramming may be facilitated by p53 inhibition, gain-of-function oncogenic mutations in p53 and oncogenic mutations in Kras-stimulated tumorigenic activity, and their roles in vivo are imperfectly understood. Hence, in the present study, the effect of direct injection of a Sendai virus (SeV) vector encoding four defined factors in vivo was studied using various backgrounds of transgenic and knockout mice, and was compared with that of direct injection of microRNAs (miRNAs) diluted with cationic lipid. The in vivo imaging data revealed transformation hot spots for p53 deficiency or conditional activation of mutant Kras, and the sizes were concordant with those in immuno-deficient NOD/SCID and uPA-NOG mice, as well as larger compared with those in the control mice. Overall, the present data on in vivo reprogramming indicated that Kras activation may facilitate the effect of cellular reprogramming in normal liver cells, and the effect of Kras activation is more apparent than that of tumor suppressor p53 deficiency. The results also revealed that immunodeficiency may increase the effect of reprogramming, presumably by blocking the immunosurveillance of transformed cells. These findings provide a rationale for further studies to develop a therapeutic approach involving direct in vivo reprogramming.

9.
Stem Cells Transl Med ; 2(8): 558-66, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23847002

RESUMEN

The discovery of induced pluripotent stem cells (iPSCs) holds great promise for regenerative medicine since it is possible to produce patient-specific pluripotent stem cells from affected individuals for potential autologous treatment. Using nonintegrating cytoplasmic Sendai viral vectors, we generated iPSCs efficiently from adult mobilized CD34⁺ and peripheral blood mononuclear cells. After 5-8 passages, the Sendai viral genome could not be detected by real-time quantitative reverse transcription-polymerase chain reaction. Using the spin embryoid body method, we showed that these blood cell-derived iPSCs could efficiently be differentiated into hematopoietic stem and progenitor cells without the need of coculture with either mouse or human stromal cells. We obtained up to 40% CD34⁺ of which ~25% were CD34⁺/CD43⁺ hematopoietic precursors that could readily be differentiated into mature blood cells. Our study demonstrated a reproducible protocol for reprogramming blood cells into transgene-free iPSCs by the Sendai viral vector method. Maintenance of the genomic integrity of iPSCs without integration of exogenous DNA should allow the development of therapeutic-grade stem cells for regenerative medicine.


Asunto(s)
Células Sanguíneas/citología , Reprogramación Celular , Vectores Genéticos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Virus Sendai/genética , Factores de Transcripción/metabolismo , Adulto , Animales , Antígenos CD34/metabolismo , Diferenciación Celular/genética , Reprogramación Celular/genética , Femenino , Perfilación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Transgenes/genética
10.
J Neurooncol ; 106(3): 519-29, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21904957

RESUMEN

To identify therapeutic molecular targets for glioma, we performed modified serological identification of antigens by recombinant complementary DNA (cDNA) expression cloning using sera from a mouse glioma model. Two clones, kinesin family member 23 (Kif23) and structural maintenance of chromosomes 4 (Smc4), were identified as antigens through immunological reaction with sera from mice harboring synergic GL261 mouse glioma and intratumoral inoculation with a mutant herpes simplex virus. The human Kif23 homolog KIF23 is a nuclear protein that localizes to the interzone of mitotic spindles, acting as a plus-end-directed motor enzyme that moves antiparallel microtubules in vitro. Expression analysis revealed a higher level of KIF23 expression in glioma tissues than in normal brain tissue. The introduction of small interfering RNA (siRNA) targeting KIF23 into two different glioma cell lines, U87MG and SF126, downregulated KIF23 expression, which significantly suppressed glioma cell proliferation in vitro. KIF23 siRNA-treated glioma cells exhibited larger cell bodies with two or more nuclei compared with control cells. In vivo analysis using mouse xenograft showed that KIF23 siRNA/DNA chimera-treated tumors were significantly smaller than tumors treated with control siRNA/DNA chimera. Taken together, our results indicate that downregulation of KIF23 decreases proliferation of glioma cells and that KIF23 may be a novel therapeutic target in malignant glioma.


Asunto(s)
Neoplasias Encefálicas/patología , Proliferación Celular , Regulación hacia Abajo/fisiología , Glioma/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Antígeno Ki-67/metabolismo , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Invasividad Neoplásica , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Simplexvirus/inmunología , Factores de Tiempo , Transfección , Trasplante Heterólogo/métodos
11.
Nature ; 478(7369): 391-4, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21993621

RESUMEN

Human induced pluripotent stem cells (iPSCs) represent a unique opportunity for regenerative medicine because they offer the prospect of generating unlimited quantities of cells for autologous transplantation, with potential application in treatments for a broad range of disorders. However, the use of human iPSCs in the context of genetically inherited human disease will require the correction of disease-causing mutations in a manner that is fully compatible with clinical applications. The methods currently available, such as homologous recombination, lack the necessary efficiency and also leave residual sequences in the targeted genome. Therefore, the development of new approaches to edit the mammalian genome is a prerequisite to delivering the clinical promise of human iPSCs. Here we show that a combination of zinc finger nucleases (ZFNs) and piggyBac technology in human iPSCs can achieve biallelic correction of a point mutation (Glu342Lys) in the α(1)-antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for α(1)-antitrypsin deficiency. Genetic correction of human iPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene-targeting technology that is currently available and crucially prevents contamination of the host genome with residual non-human sequences. Our results provide the first proof of principle, to our knowledge, for the potential of combining human iPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Reparación del Gen Blanco , Deficiencia de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/genética , Animales , Línea Celular , Elementos Transponibles de ADN/genética , Hepatocitos/metabolismo , Hepatocitos/trasplante , Humanos , Hígado/citología , Ratones , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Albúmina Sérica Humana , Factores de Tiempo , alfa 1-Antitripsina/metabolismo
12.
J Stem Cells ; 6(3): 101-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-23264996

RESUMEN

We report that iPS cells can be safely and effectively generated from fresh human cord blood (CB) cells with Sendai virus (SeV) vector carrying reprogramming factors OCT3/4, SOX2, KLF4, and c-MYC. The SeV vector is a single strand RNA virus having no DNA phase, and selectively infects the freshly isolated CD34+ CD45low+ fraction of CB cells corresponding to hematopoietic progenitors. Approximately twenty ES cell-like colonies emerged from 1 x 104 freshly isolated CD34+ CB cells around 18 days after SeV infection and were selected for passage to reduce the frequency of the remaining SeV-infected cells. The complete elimination of viral constructs was confirmed after several passages by immunostaining with monoclonal antibody against hemagglutinin-neuraminidase (HN) and by RT-PCR analysis. Five ES cell-like clones were selected to examine their in vitro potential for three germ layer differentiation and their capacity for teratoma formation. Generation of non-integrating Sendai virus (SeV) iPS cells from CB cells may be an important step to provide allogeneic iPS cell-derived therapy in the future.


Asunto(s)
Sangre Fetal/citología , Células Madre Hematopoyéticas/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Separación Celular , Células Cultivadas , Reprogramación Celular , Técnicas de Cocultivo , Células Nutrientes , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Virus Sendai/genética , Factores de Tiempo , Transfección
14.
Int J Cancer ; 126(4): 919-29, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19728337

RESUMEN

Malignant gliomas are the most aggressive human primary brain tumors and are currently incurable. Immunotherapies have the potential to target glioma and glioma stem cells (GSCs) that are resistant to conventional therapies. We previously identified SOX6 as a human glioma antigen and demonstrated that vaccination with SOX6 DNA induced cytotoxic T lymphocytes (CTLs) specific for glioma, thereby exerting therapeutic antitumor responses in glioma-bearing mice. In this study, we attempted to identify SOX6-derived peptides as specific targets for effective and safe T-cell-mediated immunotherapy targeting SOX6-positive glioma and GSCs. In vitro stimulation with human leukocyte antigen (HLA)-A*2402 (A24)-restricted peptides, RFENLGPQL (SOX6(504)) and PYYEEQARL (SOX6(628)) or the HLA-A*0201 (A2)-restricted peptide, ALFGDQDTV (SOX6(447)) was capable of inducing SOX6 peptide-specific CTLs in peripheral blood mononuclear cells derived from healthy donors and glioma patients. These CTLs were able to lyse a majority of glioma cell lines and a GSC line derived from human glioblastoma in an HLA Class I-restricted and an antigen-dependent manner. Furthermore, peptide vaccines of SOX6(628), which was conserved in the murine SOX6 protein and expected to bind to major histocompatibility complex (MHC) H-2(d), induced CTLs specific for SOX6(628) in H-2(d) mice. Normal autologous cells from mice, in which SOX6-specific immune responses were generated, were not destroyed. These results suggest that these SOX6 peptides are potnetially immunogenic in HLA-A24 or -A2 positive glioma patients and should be considered as a promising strategy for safe and effective T-cell-based immunotherapy of patients with gliomas.


Asunto(s)
Glioma/inmunología , Antígenos HLA-A/inmunología , Antígeno HLA-A2/inmunología , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Adulto , Animales , Encéfalo/inmunología , Encéfalo/patología , Neoplasias Encefálicas/epidemiología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Supervivencia Celular/inmunología , Epítopos/inmunología , Femenino , Glioma/epidemiología , Glioma/patología , Antígenos HLA-A/genética , Antígeno HLA-A24 , Antígenos HLA-B/genética , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Estados Unidos/epidemiología , Adulto Joven
15.
Artículo en Inglés | MEDLINE | ID: mdl-19838014

RESUMEN

Induced pluripotent stem cells (iPSC) have been generated from somatic cells by introducing reprogramming factors. Integration of foreign genes into the host genome is a technical hurdle for the clinical application. Here, we show that Sendai virus (SeV), an RNA virus and carries no risk of altering host genome, is an efficient solution for generating safe iPSC. Sendai-viral human iPSC expressed pluripotency genes, showed demethylation characteristic of reprogrammed cells. SeV-derived transgenes were decreased during cell division. Moreover, viruses were able to be easily removed by antibody-mediated negative selection utilizing cell surface marker HN that is expressed on SeV-infected cells. Viral-free iPSC differentiated to mature cells of the three embryonic germ layers in vivo and in vitro including beating cardiomyocytes, neurons, bone and pancreatic cells. Our data demonstrated that highly-efficient, non-integrating SeV-based vector system provides a critical solution for reprogramming somatic cells and will accelerate the clinical application.


Asunto(s)
Reprogramación Celular/genética , Vectores Genéticos/genética , Genoma Humano/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Virus Sendai/genética , Transgenes/genética , Biomarcadores/metabolismo , Línea Celular Tumoral , Proliferación Celular , Reprogramación Celular/fisiología , Metilación de ADN , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Vectores Genéticos/aislamiento & purificación , Humanos , Virus Sendai/aislamiento & purificación , Transducción Genética , Integración Viral
16.
Eur J Immunol ; 35(5): 1634-42, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15816000

RESUMEN

CD72 has been reported to regulate BCR-mediated signals both positively and negatively. SHP-1 and Grb2 bind, respectively, to ITIM1 and ITIM2 of CD72. We generated transformed B cell lines with an immature phenotype following J2 virus infection of splenocytes from CD72(-/-) and wild-type (Wt) mice. The transformed lines were infected with retroviral vectors carrying Tyr (Y) to Phe (F) substitutions in the ITIM sequences (ITIM1 mutated: Y7/F; ITIM2 mutated: Y39/F; and both ITIM mutated: Y7,39/F). Cross-linking of the BCR induced growth inhibition in transfectants expressing Wt CD72, but this response was less sensitive in transfectants with Y7,39/F. The Y7/F transfectants demonstrated the least sensitive response. We were not able to obtain transfectants with Y39/F, suggesting that CD72 associated with SHP-1, but not with Grb2, delivers a strong negative signal. Pre-ligation of CD72, which induces dephosphorylation of the molecule, partially rescued the Wt transfectants from growth inhibition, leading to a growth response profile similar to that of Y7,39/F transfectants. These results suggest that ITIM1/SHP-1 delivers a very strong negative signal that is down-modulated by signals through ITIM2/Grb2, leading to delivery of an attenuated negative signal. Thus, pre-ligation of CD72 results in the manifestation of an ostensible positive signal.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Proliferación Celular , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos B/genética , Citometría de Flujo , Proteína Adaptadora GRB2 , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/inmunología , Proteínas Tirosina Fosfatasas/metabolismo , Transfección
17.
Microbiol Immunol ; 48(1): 59-66, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14734859

RESUMEN

Engagement of mIgM induces G1 arrest and apoptosis in immature B cells. The biochemical mechanism(s) regulating the cell death process are poorly understood. Cross-linking of CD72 (a B cell co-receptor) with anti-CD72 antibody was shown to protect B cells from apoptosis. We investigated the molecular mechanism involved in apoptosis preventing signaling mediated by CD72 ligation using a derivative (WEHIdelta) of the WEHI231 cell line which is representative of immature B cells. Apoptotic WEHIdelta cells following cross-linking of mIgM demonstrate a dramatic loss of c-Myc protein after transient up-regulation. In contrast, pre-ligation of CD72 was able to sustain c-Myc expression after transient up-regulation. Cross-linking of mIgM of WEHIdelta cells causes accumulation of the Cdk inhibitor, p27(Kip1). CD72 pre-ligation was shown to inhibit the accumulation of p27(Kip1) protein. Moreover, NF-kappaB activity was not suppressed in WEHIdelta cells after mIgM cross-linking when the cells were pre-treated with anti-CD72 antibody. These results strongly suggest that the apoptosis preventing signal evoked by CD72 ligation is delivered through the pathway of NF-kappaB, c-Myc, p27(Kip1) and cyclin.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Apoptosis , Linfocitos B/fisiología , Proteínas de Ciclo Celular/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Proteínas Supresoras de Tumor/metabolismo , Animales , Ciclo Celular , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Fase G1 , Inmunoglobulina M/inmunología , Ratones , Transducción de Señal
18.
Biochem Biophys Res Commun ; 304(1): 67-72, 2003 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-12705885

RESUMEN

SHP-1 plays an important role in negative signaling in many cell types. For example, after BCR stimulation in apoptotic B cells, SHP-1 has been shown to be recruited to phosphorylated ITIMs present in receptors such as CD72. However, the SHP-1 substrates in the chicken B cell line, DT40, have been poorly undefined. To identify SHP-1 substrates in DT40, we used a trapping mutant SHP-1 C/S (a catalytically inactive form). BCR stimulation induced hyper-phosphorylation of 230 kDa protein in C/S transfectants. MALDI-TOF/MS analysis revealed that this was myosin carrying ITIM. SHP-1 was shown to bind to this ITIM in synthetic peptide binding experiment. Thus, myosin is a direct SHP-1 substrate in B cells. The results suggest that SHP-1 plays a critical role in the reorganization of cytoskeletal architecture mediated via BCR stimulation.


Asunto(s)
Linfocitos B/enzimología , Miosinas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Linfocitos B/inmunología , Sitios de Unión , Pollos , Inmunoglobulina M/inmunología , Péptidos y Proteínas de Señalización Intracelular , Mutación , Miosinas/química , Fosforilación , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/genética , Receptores de Antígenos de Linfocitos B/inmunología , Células Tumorales Cultivadas
19.
J Immunol ; 170(7): 3762-8, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12646642

RESUMEN

Src homology protein 1 (SHP-1) plays an important role in B cell Ag receptor (BCR) differentiation, proliferation, survival, and apoptosis. After BCR stimulation in apoptotic cells, SHP-1 has been shown to be recruited to phosphorylated immunoreceptor tyrosine-based inhibitory motifs present in receptors such as CD22 and CD72. However, the substrates of SHP-1 in the chicken B cell line, DT40, have remained undefined. To identify SHP-1 substrates in DT40, we used a trapping mutant, SHP-1 C/S (a catalytically inactive form). Cross-linking of BCR induced hyperphosphorylation of approximately 44-kDa protein in C/S transfectants. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis revealed that this was actin (cytoplasmic type 5) carrying three immunoreceptor tyrosine-based inhibitory motif-like sequences. SHP-1 was shown to bind to one of these sequences in synthetic peptide binding experiment. Thus, actin is a direct SHP-1 substrate. Furthermore, more SHP-1 molecules translocate into lipid rafts, and their association with actin was increased after BCR stimulation. In C/S transfectants, actin polymerization induced by membrane IgM ligation was sustained to a greater extent for a longer time compared with wild-type transfectants. Therefore, actin dephosphorylation by SHP-1 is essential for actin depolymerization after BCR stimulation. Our data suggest that SHP-1 plays a pivotal role in reorganization of cytoskeletal architecture inducing actin dephosphorylation. These results clearly demonstrate the direct interaction of SHP-1 with actin.


Asunto(s)
Actinas/metabolismo , Reactivos de Enlaces Cruzados/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Tirosina/metabolismo , Actinas/genética , Actinas/aislamiento & purificación , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Pollos , Cisteína/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Polímeros/metabolismo , Unión Proteica/genética , Transporte de Proteínas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/genética , Serina/genética , Especificidad por Sustrato/genética , Transfección , Tirosina/genética , Dominios Homologos src/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA