Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Methods ; 6(10): 733-5, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19718031

RESUMO

We developed a method for somatic cell nuclear transfer in zebrafish using laser-ablated metaphase II eggs as recipients, the micropyle for transfer of the nucleus and an egg activation protocol after nuclear reconstruction. We produced clones from cells of both embryonic and adult origins, although the latter did not give rise to live adult clones.


Assuntos
Engenharia Genética/métodos , Células Híbridas/transplante , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/genética , Animais , Técnicas de Transferência Nuclear
2.
Nat Med ; 6(3): 332-6, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10700237

RESUMO

Reports of spontaneous regressions of metastases and the demonstration of tumor-reactive cytotoxic T lymphocytes indicate the importance of the host's immune system in controlling the devastating course of metastatic renal cell carcinoma. Recent research indicates that immunization with hybrids of tumor and antigen presenting cells results in protective immunity and rejection of established tumors in various rodent models. Here, we present a hybrid cell vaccination study of 17 patients. Using electrofusion techniques, we generated hybrids of autologous tumor and allogeneic dendritic cells that presented antigens expressed by the tumor in concert with the co-stimulating capabilities of dendritic cells. After vaccination, and with a mean follow-up time of 13 months, four patients completely rejected all metastatic tumor lesions, one presented a 'mixed response', and two had a tumor mass reduction of greater 50%. We also demonstrate induction of HLA-A2-restricted cytotoxic T cells reactive with the Muc1 tumor-associated antigen and recruitment of CD8+ lymphocytes into tumor challenge sites. Our data indicate that hybrid cell vaccination is a safe and effective therapy for renal cell carcinoma and may provide a broadly applicable strategy for other malignancies with unknown antigens.


Assuntos
Vacinas Anticâncer , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/terapia , Células Dendríticas/imunologia , Células Híbridas/imunologia , Neoplasias Renais/terapia , Idoso , Idoso de 80 Anos ou mais , Vacinas Anticâncer/efeitos adversos , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Citotoxicidade Imunológica , Células Dendríticas/transplante , Humanos , Células Híbridas/transplante , Interferon gama/sangue , Neoplasias Renais/imunologia , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Linfócitos/imunologia , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Análise de Sobrevida , Fatores de Tempo , Tomografia Computadorizada por Raios X , Transplante Autólogo , Transplante Homólogo , Resultado do Tratamento
3.
Stem Cells Dev ; 30(4): 190-202, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33349121

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal X-linked disorder caused by mutations in dystrophin gene. Currently, there is no cure for DMD. Cell therapies are challenged by limited engraftment and rejection. Thus, more effective and safer therapeutic approaches are needed for DMD. We previously reported increased dystrophin expression correlating with improved function after transplantation of dystrophin expressing chimeric (DEC) cells of myoblast origin in the mdx mouse models of DMD. This study established new DEC cell line of myoblasts and mesenchymal stem cells (MSC) origin and tested its efficacy and therapeutic potential in mdx/scid mouse model of DMD. Fifteen ex vivo cell fusions of allogenic human myoblast [normal myoblasts (MBN)] and normal human bone marrow-derived MSC (MSCN) from normal donors were performed using polyethylene glycol. Flow cytometry, confocal microscopy, polymerase chain reaction (PCR)-short tandem repeats, polymerase chain reaction-reverse sequence-specific oligonucleotide probe assessed chimeric state of fused MBN/MSCN DEC cells, whereas Comet assay assessed fusion procedure safety testing genotoxicity. Immunofluorescence and real-time PCR assessed dystrophin expression and myogenic differentiation. Mixed lymphocyte reaction (MLR) evaluated DEC's immunogenicity. To test MBN/MSCN DEC efficacy in vivo, gastrocnemius muscle of mdx/scid mice were injected with vehicle (n = 12), nonfused MBN and MSCN (n = 9, 0.25 × 106/each) or MBN/MSCN DEC (n = 9, 0.5 × 106). Animals were evaluated for 90 days using ex vivo and in vivo muscle strength tests. Histology and immunofluorescence staining assessed dystrophin expression, centrally nucleated fibers and scar tissue formation. Post-fusion, MBN/MSCN DEC chimeric state, myogenic differentiation, and dystrophin expression were confirmed. MLR reveled reduced DEC's immune response compared with controls (P < 0.05). At 90 days post-DEC transplant, increase in dystrophin expression (20.26% ± 2.5%, P < 0.05) correlated with improved muscle strength and function in mdx/scid mice. The created human MBN/MSCN DEC cell line introduces novel therapeutic approach combining myogenic and immunomodulatory properties of MB and MSC, and as such may open a universal approach for muscle regeneration in DMD.


Assuntos
Distrofina/genética , Células Híbridas/transplante , Células-Tronco Mesenquimais/metabolismo , Distrofia Muscular de Duchenne/terapia , Mioblastos/metabolismo , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular/genética , Fusão Celular , Células Cultivadas , Modelos Animais de Doenças , Distrofina/metabolismo , Expressão Gênica , Humanos , Células Híbridas/citologia , Células Híbridas/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos SCID , Músculo Esquelético/citologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Mioblastos/citologia , Transplante Heterólogo
4.
Int J Cancer ; 126(5): 1206-15, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19653275

RESUMO

The prognosis of patients with glioblastoma multiforme (GBM) is generally poor after surgical tumor resection. With the aim of developing new adjuvant therapeutic strategies, we have investigated primary neural stem/progenitor cells (NSPC) in co-cultures with glioma cells, and in a model of gene therapy on aggressively growing malignant glioma. NSPC exhibited tropism towards medium conditioned by glioma cells, and in adherent low-cell density co-culture, were attracted to, and fused with, tumor cells. Similarly, within 24-48 hr of co-culture in suspension, NSPC-tumor hybrids were observed, representing 2-3% of the total cell population. NSPC were then coinjected into mouse brain with GBM cells, employing NSPC expressing cyclophosphamide (CPA)-activating enzyme cytochrome p450 2B6 (CYP2B6), which catalyzes CPA prodrug transformation into membrane diffusible DNA-alkylating metabolites. Upon CPA administration, NSPC containing CYP2B6 elicited substantial impairment of tumor growth. When implanted intracerebrally at a distant site from the tumor, gene-engineered NSPC specifically targeted GBM grafts, after traveling through brain parenchyma, and hindered tumor growth through local activation of CPA. Directed migration of primary NSPC corresponded closely with intracerebral and tumoral pattern of expression of vascular endothelial growth factor, which is a motility factor for NSPC. Overall, these findings indicate that therapeutic gene delivery mediated by primary NSPC is a potentially valid strategy for treatment of high-grade gliomas.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Células Híbridas/transplante , Neurônios/citologia , Células-Tronco/citologia , Animais , Antineoplásicos Alquilantes/farmacologia , Hidrocarboneto de Aril Hidroxilases/genética , Diferenciação Celular , Movimento Celular , Técnicas de Cocultura , Ciclofosfamida/farmacologia , Citocromo P-450 CYP2B6 , Engenharia Genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases N-Desmetilantes/genética , Transdução Genética
5.
Scand J Gastroenterol ; 45(6): 707-13, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20205622

RESUMO

OBJECTIVE: Colorectal cancer (CRC) is one of the most common malignancies in the western world. More than 60% among patients will develop liver metastases. Although surgical resection is the first choice worldwide, at this point an effective approach for the treatment of patients with liver metastasis and cancer recurrence postoperation has not yet been found. The aim of this study is to investigate the role of the allogeneic dendritomas from fusion of DCs and metastatic colon cancer cells in the activation of anti-tumor immunity against colorectal cancer liver metastases. MATERIAL AND METHODS: Hybrids were generated by fused allogeneic human peripheral blood dendritic cells with metastatic colon cancer SW620 cells using 50% polyethylene glycol (PEG). Induction of immune responses was assessed by ex vivo ELISPOT assays. A murine model of CRC liver metastasis was used by intrasplenic injection. The validity of the vaccine was observed by Vaccination CRC liver metastasis murine model with DC/tumor hybrids. RESULTS: The hybrids highly express the major molecules of DCs and tumor cells. The number of hybrids pulsed CTL secreting IFN-gamma was significantly higher when compared to the DC controls (p < 0.01). In a therapeutic setting mice vaccinated with in vitro cultured hybrids produced strong cellular immune responses and significant inhibition of tumor growth, compared to sham vaccinated controls. CONCLUSIONS: Vaccination with hybrids can induces strong cellular responses and significant protection from challenge in SCID mouse metastatic CRC model.


Assuntos
Neoplasias Colorretais/terapia , Células Dendríticas/transplante , Células Híbridas/transplante , Imunidade Celular , Neoplasias Hepáticas/terapia , Vacinação/métodos , Animais , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Células Dendríticas/imunologia , Humanos , Células Híbridas/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Camundongos SCID , Neoplasias Experimentais , Transplante Homólogo , Resultado do Tratamento , Células Tumorais Cultivadas
6.
J Cell Biol ; 103(3): 795-805, 1986 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3745269

RESUMO

Fusion of mouse L929 cytoplasts with human peripheral blood lymphocytes induced lymphocyte proliferation that gave rise to lymphoid cell lines of B and T cell origin with unlimited growth potential. The immortalized cell lines were routinely grown in standard medium supplemented with fetal calf serum. Furthermore these cell lines could be propagated in chemically defined serum-free media. Each establishment of lymphoid cell lines was preceded by a proliferation phase 2 wk after cytoplast/cell fusion, which appears to be a necessary step in the immortalization process. The immortalized cells have a nearly normal human karyotype, do not form colonies in soft agar medium, and are not tumorigenic in nude mice. Cloned B cell lines produced human immunoglobulins of heavy and light chain types. No cross-reaction with DNA of herpes simplex virus, human cytomegalovirus, human T cell leukemia/lymphoma virus I and II, or polyoma virus was detected in the genome of immortalized cell lines by Southern blot hybridization. Furthermore B and T cell lines were established that appear to be free of Epstein-Barr virus genome.


Assuntos
Células Híbridas/fisiologia , Células L/fisiologia , Linfócitos/fisiologia , Animais , Fusão Celular , Sobrevivência Celular , Citoplasma/fisiologia , DNA Viral/análise , Humanos , Células Híbridas/imunologia , Células Híbridas/transplante , Imunoglobulinas/biossíntese , Camundongos , Camundongos Nus , Neoplasias Experimentais
7.
Cancer Res ; 65(5): 1655-63, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15753359

RESUMO

The role of mitochondrial dysfunction in cancer has been a subject of great interest and much ongoing investigation. Although most cancer cells harbor somatic mutations in mitochondrial DNA (mtDNA), the question of whether such mutations contribute to the promotion of carcinomas remains unsolved. Here we used trans-mitochondrial hybrids (cybrids) containing a common HeLa nucleus and mtDNA of interest to compare the role of mtDNA against the common nuclear background. We constructed cybrids with or without a homoplasmic pathogenic point mutation at nucleotide position 8,993 or 9,176 in the mtDNA ATP synthase subunit 6 gene (MTATP6) derived from patients with mitochondrial encephalomyopathy. When the cybrids were transplanted into nude mice, the MTATP6 mutations conferred an advantage in the early stage of tumor growth. The mutant cybrids also increased faster than wild type in culture. To complement the mtDNA mutations, we transfected a wild-type nuclear version of MTATP, whose codons were converted to the universal genetic codes containing a mitochondrial target sequence, into the nucleus of cybrids carrying mutant MTATP6. The restoration of MTATP slowed down the growth of tumor in transplantation. Conversely, expression of a mutant nuclear version of MTATP6 in the wild-type cybrids declined respiration and accelerated the tumor growth. These findings showed that the advantage in tumor growth depended upon the MTATP6 function but was not due to secondary nuclear mutations caused by the mutant mitochondria. Because apoptosis occurred less frequently in the mutant versus wild-type cybrids in cultures and tumors, the pathogenic mtDNA mutations seem to promote tumors by preventing apoptosis.


Assuntos
Adenosina Trifosfatases/genética , Apoptose , DNA Mitocondrial/genética , Mitocôndrias/genética , Neoplasias Experimentais/etiologia , Mutação Puntual , Adenosina Trifosfatases/metabolismo , Adolescente , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Feminino , Fibroblastos , Genoma , Células HeLa , Humanos , Células Híbridas/transplante , Lactente , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Encefalomiopatias Mitocondriais/metabolismo , Encefalomiopatias Mitocondriais/patologia , ATPases Mitocondriais Próton-Translocadoras , Neoplasias Experimentais/metabolismo , Oxigênio/metabolismo , Pele/metabolismo
8.
Int J Oncol ; 28(3): 731-6, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16465379

RESUMO

The present study demonstrates that immunization with a low dose of unmodified live myeloma tumor cells (FO) elicited tumor-specific immunity. BALB/c mice were vaccinated with 10(4) live dendritic cells (DC)-FO fusion cells or 10(3) live FO cells. 80% of vaccinated mice survived from the later challenge with 1 x 10(6) FO cells, whereas all control mice developed tumors. Additionally, vaccination with live FO cells gave no protection against the growth of Lewis lung carcinoma cells in C57BL/6 mice. Cellular immunity was found to be primarily responsible for anti-tumor responses. In an adoptive immune model, the development of myeloma was greatly reduced by transfusion of lymphocytes but not sera from mice immunized with FO. T cells from immunized mice also induced lysis of FO cells in the cytotoxic T lymphocyte (CTL) assay. After co-culture with FO, IFN-gamma released from immunized T helper cells increased >10-fold, while IL-4 remained unchanged in comparison with control T cells. These findings provided the first evidence that immunization with a low dose of unmodified live FO cells was safe to mice and capable of eliciting specific protective immunity against tumor growth.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia Adotiva/métodos , Neoplasias Experimentais/terapia , Animais , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/transplante , Relação Dose-Resposta a Droga , Feminino , Células Híbridas/imunologia , Células Híbridas/transplante , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Autólogo
9.
Int J Oncol ; 28(3): 585-93, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16465362

RESUMO

A pilot clinical trial using dendritomas, purified hybrids from the fusion of dendritic/tumor cells combined with a low dose of IL-2, in metastatic melanoma patients was conducted in order to determine its safety and potential immunological and clinical responses. Ten metastatic melanoma patients were enrolled into this study. Dendritoma vaccines were created by fusing dendritic cells stained with green fluorescent dye with irradiated autologous tumor cells stained with red fluorescent dye and purifying the hybrids using immediate fluorescent-activated cell sorting. Initial vaccine was given subcutaneously and followed by IL-2 in serially elevated doses from 3-9 million units/m2 for 5 days. Repeated vaccinations were administered without IL-2, at 3-month intervals for a maximum of 5 times. Immune reactions were measured by the increase of interferon-gamma (IFN-gamma) expressing T cells. Vaccine doses ranged from 250,000 to 1,000,000 dendritomas. There was no grade 2 or higher toxicity directly attributable to the vaccine. All patients experienced toxicity due to IL-2 administration (9-grade 2, 3-grade 3, 1-grade 4). Eight of nine evaluable patients demonstrated immunologic reactions by increased IFN-gamma expressing T cells. One patient developed partial response at 12 weeks after the first vaccine. Nine months later, this patient achieved a complete response. In addition, two patients had stable disease for 9 and 4 months, respectively; one patient had a mixed response. Our findings demonstrated that dendritoma vaccines with a low dose of IL-2 can be safely administered to patients with metastatic melanoma and induce immunological and clinical responses.


Assuntos
Células Dendríticas/imunologia , Interleucina-2/uso terapêutico , Melanoma/terapia , Neoplasias Cutâneas/terapia , Adulto , Idoso , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/citologia , Feminino , Citometria de Fluxo , Humanos , Células Híbridas/imunologia , Células Híbridas/transplante , Imunoterapia Adotiva/métodos , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Masculino , Melanoma/imunologia , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Projetos Piloto , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Resultado do Tratamento
11.
J Natl Cancer Inst ; 68(5): 841-9, 1982 May.
Artigo em Inglês | MEDLINE | ID: mdl-6951093

RESUMO

The cell line designated PIR-2 is a nonimmunogenic X-ray-induced thymoma of C57BL/6 origin that is unable to induce antitumor immunity in syngeneic lymphocytes in vitro and in mice in vivo. Fusion of PIR-2 with an allogeneic "universal fuser" A9HT (clone 3c) resulted in the establishment of a somatic cell hybrid designated A9/PIR. C57BL/6 lymphocytes sensitized in vitro with A9/PIR could lyse parental PIR-2 cells, as well as other syngeneic tumors. However, immunization of mice with the hybrid significantly enhanced PIR-2 tumor takes while it partially protected the animals against a challenge with unrelated syngeneic tumors. The results imply that somatic cell hybridization can increase the immunogenicity of an otherwise nonimmunogenic tumor. However, in view of the enhancing effects of hybrid preimmunization on parental tumor cell growth, the possible application of this approach for immunotherapy is questionable.


Assuntos
Células Híbridas/imunologia , Linfoma/imunologia , Animais , Linhagem Celular , Feminino , Células Híbridas/transplante , Imunização , Isoantígenos/análise , Linfoma/mortalidade , Linfoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Induzidas por Radiação/imunologia , Fatores de Tempo
12.
Cancer Res ; 43(5): 2240-6, 1983 May.
Artigo em Inglês | MEDLINE | ID: mdl-6831446

RESUMO

Somatic cell hybrids were generated between Chinese hamster cell lines (Cl-4 and TK 17-O) with a near-diploid number of partially abnormal chromosomes and embryonic mouse fibroblasts (BALB/c). Hybrids harboring a near-diploid, near-triploid, and near-tetraploid set of hamster chromosomes plus 22 to 30 mouse chromosomes were analyzed for the expression of the transformed or tumorigenic phenotype, respectively, indicated by their capacity to form colonies in soft agar and by tumor formation after s.c. injection into nude mice. The hybrids showed (partial) suppression of tumorigenicity and of anchorage independence. The minimum number of hybrid cells required to initiate tumor growth in nude mice was 100- to 50,000-fold higher, and the latency period was 3- to 6-fold longer in comparison with the highly tumorigenic parental hamster cells. Suppression of tumorigenicity was also found in intraspecific Chinese hamster hybrids involving tumorigenic cells (E 36-O and TK 17-O) and embryonic hamster fibroblasts. To identify those mouse chromosomes associated with suppression of tumorigenicity, we investigated the expression of mouse isozyme genes and the presence of mouse chromosomes in interspecific suppressed hybrids and their tumorigenic hybrids described previously. No single mouse chromosome, even if present in two copies, and no combination of two different mouse chromosomes was sufficient to suppress tumorigenicity in these hybrids. This conclusion is based on either the presence of these chromosomes in hybrids isolated from tumors or their absence in suppressed hybrids.


Assuntos
Fibroblastos/ultraestrutura , Células Híbridas/ultraestrutura , Neoplasias/ultraestrutura , Ágar , Animais , Divisão Celular , Linhagem Celular , Aberrações Cromossômicas , Cromossomos/ultraestrutura , Cricetinae , Cricetulus , Embrião de Mamíferos , Genótipo , Células Híbridas/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus
13.
Cancer Res ; 59(9): 2182-9, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10232606

RESUMO

Human chromosome 3p cytogenetic abnormalities and loss of heterozygosity have been observed at high frequency in the nonpapillary form of sporadic renal cell carcinoma (RCC). The von Hippel-Lindau (VHL) gene has been identified as a tumor suppressor gene for RCC at 3p25, and functional studies as well as molecular genetic and cytogenetic analyses have suggested as many as two or three additional regions of 3p that could harbor tumor suppressor genes for sporadic RCC. We have previously functionally defined a novel genetic locus nonpapillary renal carcinoma-1 (NRC-1) within chromosome 3p12, distinct from the VHL gene, that mediates tumor suppression and rapid cell death of RCC cells in vivo. We now report the suppression of tumorigenicity of RCC cells in vivo after the transfer of a defined centric 3p fragment into different histological types of RCC. Results document the functional involvement of NRC-1 in not only different cell types of RCC (i.e., clear cell, mixed granular cell/clear cell, and sarcomatoid types) but also in papillary RCC, a less frequent histological type of RCC for which chromosome 3p LOH and genetic aberrations have only rarely been observed. We also report that the tumor suppression observed in functional genetic screens was independent of the microenvironment of the tumor, further supporting a role for NRC-1 as a more general mediator of in vivo growth control. Furthermore, this report demonstrates the first functional evidence for a VHL-independent pathway to tumorigenesis in the kidney via the genetic locus NRC-1.


Assuntos
Carcinoma de Células Renais/genética , Cromossomos Humanos Par 3/genética , Genes Supressores de Tumor , Neoplasias Renais/genética , Ligases , Proteínas Quinases/fisiologia , Proteínas/fisiologia , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases , Animais , Carcinoma Papilar/genética , Carcinoma Papilar/patologia , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/patologia , Transformação Celular Neoplásica/genética , Feminino , Deleção de Genes , Teste de Complementação Genética , Humanos , Células Híbridas/transplante , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Repetições de Microssatélites , Pessoa de Meia-Idade , Quinases de Proteína Quinase Ativadas por Mitógeno , Transplante de Neoplasias , Neovascularização Patológica/genética , Proteínas Quinases/genética , Proteínas/genética , Células Tumorais Cultivadas/transplante , Proteína Supressora de Tumor Von Hippel-Lindau
14.
Circulation ; 108(7): 889-95, 2003 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12835224

RESUMO

BACKGROUND: Circulating endothelial progenitor cells (EPCs) migrate to injured vascular endothelium and differentiate into mature endothelial cells. We investigated whether transplantation of vasodilator gene-transduced EPCs ameliorates monocrotaline (MCT)-induced pulmonary hypertension in rats. METHODS AND RESULTS: We obtained EPCs from cultured human umbilical cord blood mononuclear cells and constructed plasmid DNA of adrenomedullin (AM), a potent vasodilator peptide. We used cationic gelatin to produce ionically linked DNA-gelatin complexes. Interestingly, EPCs phagocytosed plasmid DNA-gelatin complexes, which allowed nonviral, highly efficient gene transfer into EPCs. Intravenously administered EPCs were incorporated into the pulmonary vasculature of immunodeficient nude rats given MCT. Transplantation of EPCs alone modestly attenuated MCT-induced pulmonary hypertension (16% decrease in pulmonary vascular resistance). Furthermore, transplantation of AM DNA-transduced EPCs markedly ameliorated pulmonary hypertension in MCT rats (39% decrease in pulmonary vascular resistance). MCT rats transplanted with AM-expressing EPCs had a significantly higher survival rate than those given culture medium or EPCs alone. CONCLUSIONS: Umbilical cord blood-derived EPCs had a phagocytosing action that allowed nonviral, highly efficient gene transfer into EPCs. Transplantation of AM gene-transduced EPCs caused significantly greater improvement in pulmonary hypertension in MCT rats than transplantation of EPCs alone. Thus, a novel hybrid cell-gene therapy based on the phagocytosing action of EPCs may be a new therapeutic strategy for the treatment of pulmonary hypertension.


Assuntos
Endotélio Vascular/citologia , Terapia Genética/métodos , Células Híbridas/transplante , Hipertensão Pulmonar/terapia , Transplante de Células-Tronco/métodos , Adrenomedulina , Animais , Células Cultivadas , DNA/metabolismo , Modelos Animais de Doenças , Portadores de Fármacos , Sangue Fetal/citologia , Gelatina , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde , Humanos , Células Híbridas/citologia , Células Híbridas/metabolismo , Hipertensão Pulmonar/induzido quimicamente , Proteínas Luminescentes/genética , Masculino , Monocrotalina , Peptídeos/genética , Peptídeos/metabolismo , Peptídeos/farmacologia , Fagocitose , Circulação Pulmonar/efeitos dos fármacos , Ratos , Ratos Nus , Células-Tronco/citologia , Taxa de Sobrevida , Resistência Vascular/efeitos dos fármacos
15.
Biomed Pharmacother ; 59(6): 275-82, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16023826

RESUMO

This work was designed to study an alternative treatment of diabetes mellitus by using a transplant of hybrid cells obtained by the electrofusion of pancreatic islet cells from a healthy donor with dermic cells obtained from a recipient. The hybrid cells kept the capacity of insulin production, its regulation, and the natural control of glycemia, as well as the factors of histocompatibility to avoid the rejection. Four groups of four rats each were established: Group 1. Healthy animals (healthy control), Group 2. Diabetized non-treated animals (diabetic control), Group 3. Transplant recipient rats with extraction of dermic cells which were mixed with pancreatic insular cells from a healthy donor (transplant without fusion), and Group 4. Transplant recipient rats, with extraction of dermic cells which were electrofused with pancreatic insular cells from a healthy donor (transplant with fusion). For the Group 4, the cells were combined and they were submitted to dielectrophoresis conditions with an alternating current pulse of 15 s of 10 V RMS of 0.5 MHz. The fusion was made with a direct current pulse of 1 ms of 300 V. Clinical signs were registered (weight, diuresis, food and water intake), and several biochemical parameters in blood which included basal glycemia, uric acid, cholesterol, triglycerides, glutamate oxalacetate transaminase, glutamate pyruvate transaminase, urea, creatinine, insulin, glycated hemoglobin were registered. Additionally, ketone bodies and glucose were also measured in urine. All determinations were made at 30, 60, and 90 days. Animals of Group 1 maintained its parameters within the normal ranges. Rats of Group 2 presented alterations corresponding to a diabetic state in almost all the parameters measured, none of the animals showed a tendency to improve spontaneously, two of the rats died at 66 and 72 days. The Group 3 showed a clinical profile similar to the diabetic control group without improvement, only one rat died at day 33, while in the rats transplanted with fusion (Group 4) an improvement was observed on some parameters including body weight, water intake and glycemia. Although insulin concentration was under the normal range, it was higher than in the Group 3. None rat died. These results indicate that it is possible to improve the diabetic profile by the transplant of dermic cells from a diabetic animal fused with insular cells from a healthy donor in the recipient animal.


Assuntos
Derme/citologia , Diabetes Mellitus Experimental/cirurgia , Células Híbridas/transplante , Ilhotas Pancreáticas/citologia , Alanina Transaminase/metabolismo , Animais , Aspartato Aminotransferases/metabolismo , Glicemia/metabolismo , Fusão Celular/métodos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Ingestão de Líquidos/fisiologia , Ingestão de Alimentos/fisiologia , Hemoglobinas Glicadas/metabolismo , Células Híbridas/citologia , Insulina/sangue , Corpos Cetônicos/urina , Masculino , Ratos , Ratos Wistar , Aumento de Peso/fisiologia
16.
Folia Biol (Praha) ; 51(1): 19-24, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15783088

RESUMO

Utilization of vaccines generated by fusion of dendritic cells and tumour cells is a promising approach to tumour immunotherapy. We have examined the therapeutic efficacy of vaccines generated by fusion of HPV16-associated tumour cells TC-1 with syngeneic and allogeneic dendritic cells. Locally administered hybrid cells generated by fusion of MHC class I+ TC-1 cells and syngeneic DC inhibited the growth of MHC class I+ TC-1 tumours, but not the growth of MHC class I- TC-1/A9-derived tumours. The growth of TC-1 tumours was also inhibited by hybrids generated by fusion of TC-1 cells and allogeneic DC. The therapeutic efficacy was enhanced by co-administration of the vaccine with synthetic immunostimulatory ODN CpG 1826.


Assuntos
Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral/imunologia , Células Dendríticas/imunologia , Células Híbridas/imunologia , Células Híbridas/transplante , Imunoterapia/métodos , Adjuvantes Imunológicos/farmacologia , Animais , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral/transplante , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/citologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Proteínas Oncogênicas Virais/genética , Proteínas Repressoras/genética , Resultado do Tratamento
17.
Methods Mol Biol ; 1313: 107-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25947659

RESUMO

Auxiliary use of mesenchymal stem/stromal cells (MSCs) to islet transplantation is shown to enhance efficacy. We hypothesized cell fusion of islet cells and MSCs may provide a new cell source with robustness of MSCs and islet cell function. We succeeded electrofusion between dispersed islet cells and MSCs in rats and fused cells sustained beta-cell function in vitro and in vivo, suggesting their possibility of therapeutic application. Here, we describe our method of cell fusion that enabled us to fuse islet cells to MSCs.


Assuntos
Fusão Celular , Terapia Baseada em Transplante de Células e Tecidos , Células Híbridas/citologia , Células Híbridas/transplante , Ilhotas Pancreáticas/citologia , Células-Tronco Mesenquimais/citologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ratos
18.
Oncol Rep ; 34(4): 2022-30, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26238144

RESUMO

The ability of tumor cells to autonomously generate tumor vessels has received considerable attention in recent years. However, the degree of autonomy is relative. Meanwhile, the effect of bone marrow-derived mesenchymal stem cells (BMSCs) on tumor neovascularization has not been fully elucidated. The present study aimed to illuminate whether cell fusion between glioma stem cells and BMSC is involved in glioma neovascularization. BMSCs were isolated from transgenic nude mice, of which all nucleated cells express green fluorescent protein (GFP). The immunophenotype and multilineage differentiation potential of BMSC were confirmed. SU3 glioma stem/progenitor cells were transfected with red fluorescent protein (SU3-RFP cells). In a co-culture system of BMSC-GFP and SU3-RFP, RFP+/GFP+ cells were detected and isolated by dual colors using FACS. The angiogenic effect of RFP+/GFP+ cells was determined in vivo and in vitro. Flow cytometry analysis showed that BMSC expressed high levels of CD105, C44, and very low levels of CD45 and CD11b. When co-cultured with SU3-RFP, 73.8% of cells co-expressing RFP and GFP were identified as fused cells in the 5th generation. The fused cells exhibited tube formation ability in vitro and could give rise to a solid tumor and form tumor blood vessels in vivo. In the dual-color orthotopic model of transplantable xenograft glioma, yellow vessel-like structures that expressed CD105, RFP and GFP were identified as de novo-formed vessels derived from the fused cells. The yellow vessels observed in the tumor-bearing mice directly arose from the fusion of BMSCs and SU3-RFP cells. Thus, cell fusion is one of the driving factors for tumor neovascularization.


Assuntos
Glioma/irrigação sanguínea , Glioma/patologia , Células Híbridas/transplante , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Neoplásicas/fisiologia , Animais , Fusão Celular , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Glioma/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Camundongos Transgênicos , Células-Tronco Neoplásicas/metabolismo , Transplante Heterólogo
19.
Clin Exp Metastasis ; 16(4): 299-312, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9626809

RESUMO

Studies were conducted on the hypothesis that melanoma metastasis might be initiated through the generation of hybrids comprised of cells of the primary tumor and tumor-infiltrating leukocytes. Fusion hybrids were generated in vitro between weakly metastatic Cloudman S91 mouse melanoma cells and normal mouse or human macrophages. Hybrids were implanted s.c. in the tail and mice were monitored for metastases. Controls included parental S91 cells, autologous S91 x S91 hybrids, and B16F10 melanoma cells. Of 35 hybrids tested, most were more aggressive than the parental melanoma cells, producing metastases sooner and in more mice. A striking characteristic was heterogeneity amongst hybrids, with some lines producing no metastases and others producing metastases in up to 80% of mice. With few exceptions, hybrids with the highest metastatic potential also had the highest basal melanin content whereas those with the lowest metastatic potential were basally amelanotic, as were the parental melanoma cells. A spontaneous in vivo supermelanotic hybrid between an S91 tumor cell and DBA/2J host cell was one of the most metastatic lines. Hybrids with the highest metastatic potential also exhibited markedly higher chemotaxis to fibroblast-conditioned media. Histologically, the metastatic hybrids demonstrated vascular invasion and spread to distant organs similar to that of metastatic melanomas in mice and humans. Thus previous findings of enhanced metastasis in leukocyte x lymphoma hybrids can now be extended to include leukocyte x melanoma hybrids. Whether such hybridization is a natural cause of metastasis in vivo remains to be determined; however the fusion hybrids with genetically-matched parents described herein so closely resembled naturally-occurring metastatic melanoma cells that they could serve as useful new models for studies of this complex and deadly phenomenon.


Assuntos
Células Híbridas/transplante , Macrófagos/transplante , Melanoma Experimental/secundário , Animais , Progressão da Doença , Feminino , Humanos , Células Híbridas/metabolismo , Células Híbridas/patologia , Macrófagos/metabolismo , Melaninas/biossíntese , Melanoma Experimental/genética , Melanoma Experimental/mortalidade , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Transplante de Neoplasias
20.
Clin Exp Metastasis ; 17(7): 601-7, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10845559

RESUMO

The incidence of melanoma continues to increase at a rapid rate. As for most cancers, it is melanoma metastases, rather than the primary malignancy, that is the principal cause of death. We previously showed that the introduction of a normal copy of chromosome 6 into the metastatic human melanoma cell line C8161 suppresses metastasis at a step subsequent to tumor cells entering the bloodstream. To better define the step(s) in metastasis blocked by the addition of chromosome 6 we engineered cells that constitutively express green fluorescent protein (GFP). When these tagged, chromosome 6 hybrid cells were injected intravenously into athymic mice, grossly detectable metastases did not form. However, fluorescence microscopy revealed micro-metastases (single cells or clusters of <10 cells) in the lungs, suggesting that these cells lodged in the lungs but failed to proliferate. Cells isolated from lung up to 60 days post-injection grew in culture and/or formed tumors when injected into the skin, indicating that they were still viable, but dormant. This result implies that the gene(s) on chromosome 6 interfere specifically with growth regulatory response in the lung, but not in the skin. Thus, the gene(s) responsible for metastasis suppression represents a new class of metastasis inhibitors acting at the final stages of the metastatic cascade--that is, affecting the ability of the cells to survive and proliferate at a specific secondary site.


Assuntos
Cromossomos Humanos Par 6/genética , Técnicas de Transferência de Genes , Genes Supressores de Tumor , Neoplasias Pulmonares/secundário , Melanoma/patologia , Metástase Neoplásica/genética , Células Neoplásicas Circulantes , Animais , Divisão Celular , Sobrevivência Celular , Feminino , Genes Reporter , Proteínas de Fluorescência Verde , Humanos , Células Híbridas/transplante , Injeções Intradérmicas , Injeções Intravenosas , Proteínas Luminescentes/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Melanoma/genética , Melanoma Experimental , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Metástase Neoplásica/prevenção & controle , Transplante de Neoplasias , Especificidade de Órgãos , Proteínas Recombinantes de Fusão/análise , Neoplasias Cutâneas/secundário , Transplante Heterólogo , Células Tumorais Cultivadas/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA