Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cell Rep Med ; 5(3): 101449, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38508141

RESUMEN

Tissue regeneration following an injury requires dynamic cell-state transitions that allow for establishing the cell identities required for the restoration of tissue homeostasis and function. Here, we present a biochemical intervention that induces an intermediate cell state mirroring a transition identified during normal differentiation of myoblasts and other multipotent and pluripotent cells to mature cells. When applied in somatic differentiated cells, the intervention, composed of one-carbon metabolites, reduces some dedifferentiation markers without losing the lineage identity, thus inducing limited reprogramming into a more flexible cell state. Moreover, the intervention enabled accelerated repair after muscle injury in young and aged mice. Overall, our study uncovers a conserved biochemical transitional phase that enhances cellular plasticity in vivo and hints at potential and scalable biochemical interventions of use in regenerative medicine and rejuvenation interventions that may be more tractable than genetic ones.


Asunto(s)
Músculos , Mioblastos , Ratones , Animales , Diferenciación Celular , Mioblastos/metabolismo
4.
J Clin Biochem Nutr ; 74(1): 57-62, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38292119

RESUMEN

Immune checkpoint therapy has been shown to be an effective therapy for many types of tumors. Much attention has been paid to the development of an effector target would be helpful for immune checkpoint therapy. Genistein has been shown to have an anti-tumor effect both in vitro and in vivo. In this study, we examined the effect of genistein on immune checkpoint blockade therapy against B16F1 melanoma tumors. Mice treated with genistein or anti-programmed death (PD)-1 antibody showed a significant decrease in tumor growth. However, treatment with genistein had no effect on or attenuated the efficacy of immune checkpoint therapy. The percentages of T cell receptor (TCR)ß+CD4+ and TCRß+CD8+ cells and the concentrations of interferon-γ and tumor necrosis factor-α in tumor tissue were not different among the experimental groups. A significant difference was also not found in microbe composition. Interestingly, a high expression level of PD-ligand (L)1 closely reflected the outcome of therapy by genistein or anti-PD-1 antibody. The study showed that a combination of genistein treatment does not improve the effect of immune blockade therapy. It also showed that a high PD-L1 expression level in tumors is a good prediction maker for the outcome of tumor therapy.

5.
Biosci Biotechnol Biochem ; 87(1): 90-98, 2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36352466

RESUMEN

We have previously reported that the dipeptide Phe-Pro affects lipid metabolism in vivo and in vitro, but very little is known regarding the mechanism of action of Phe-Pro after it is absorbed by the intestines via PepT1. In this study, we administered a single oral dose of Phe-Pro to rats and quantified its concentration in the portal plasma using LC-TOF/MS analysis. Additionally, the physiological blood concentration of Phe-Pro was added to the lipid accumulation model of HepG2 cells to decrease intracellular cholesterol and increase the expression of CYP7A1 and PPARα mRNA levels. Moreover, we analyzed the binding of PPARα and Phe-Pro using AlphaFold2. We found that Phe-Pro is a ligand for PPARα. To the best of our knowledge, this is the first study that shows Phe-Pro to be present in the portal plasma. We found for the first time that Phe-Pro ameliorated cholesterol metabolism in HepG2 cells.


Asunto(s)
PPAR alfa , Fenilalanina , Ratas , Animales , Humanos , Células Hep G2 , PPAR alfa/metabolismo , Fenilalanina/farmacología , Fenilalanina/metabolismo , Prolina/farmacología , Prolina/metabolismo , Colesterol/metabolismo , Metabolismo de los Lípidos
6.
Sci Transl Med ; 14(657): eabl6057, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947677

RESUMEN

Constitutive heterochromatin is responsible for genome repression of DNA enriched in repetitive sequences, telomeres, and centromeres. During physiological and pathological premature aging, heterochromatin homeostasis is profoundly compromised. Here, we showed that LINE-1 (Long Interspersed Nuclear Element-1; L1) RNA accumulation was an early event in both typical and atypical human progeroid syndromes. L1 RNA negatively regulated the enzymatic activity of the histone-lysine N-methyltransferase SUV39H1 (suppression of variegation 3-9 homolog 1), resulting in heterochromatin loss and onset of senescent phenotypes in vitro. Depletion of L1 RNA in dermal fibroblast cells from patients with different progeroid syndromes using specific antisense oligonucleotides (ASOs) restored heterochromatin histone 3 lysine 9 and histone 3 lysine 27 trimethylation marks, reversed DNA methylation age, and counteracted the expression of senescence-associated secretory phenotype genes such as p16, p21, activating transcription factor 3 (ATF3), matrix metallopeptidase 13 (MMP13), interleukin 1a (IL1a), BTG anti-proliferation factor 2 (BTG2), and growth arrest and DNA damage inducible beta (GADD45b). Moreover, systemic delivery of ASOs rescued the histophysiology of tissues and increased the life span of a Hutchinson-Gilford progeria syndrome mouse model. Transcriptional profiling of human and mouse samples after L1 RNA depletion demonstrated that pathways associated with nuclear chromatin organization, cell proliferation, and transcription regulation were enriched. Similarly, pathways associated with aging, inflammatory response, innate immune response, and DNA damage were down-regulated. Our results highlight the role of L1 RNA in heterochromatin homeostasis in progeroid syndromes and identify a possible therapeutic approach to treat premature aging and related syndromes.


Asunto(s)
Envejecimiento Prematuro , Síndrome de Cockayne , Proteínas Inmediatas-Precoces , Progeria , Envejecimiento Prematuro/genética , Animales , Antígenos de Diferenciación , Heterocromatina , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Elementos de Nucleótido Esparcido Largo , Lisina/metabolismo , Ratones , Fenotipo , Progeria/genética , ARN , Telómero/genética , Proteínas Supresoras de Tumor/genética
7.
Cell Rep ; 39(4): 110730, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476977

RESUMEN

Mammals have limited regenerative capacity, whereas some vertebrates, like fish and salamanders, are able to regenerate their organs efficiently. The regeneration in these species depends on cell dedifferentiation followed by proliferation. We generate a mouse model that enables the inducible expression of the four Yamanaka factors (Oct-3/4, Sox2, Klf4, and c-Myc, or 4F) specifically in hepatocytes. Transient in vivo 4F expression induces partial reprogramming of adult hepatocytes to a progenitor state and concomitantly increases cell proliferation. This is indicated by reduced expression of differentiated hepatic-lineage markers, an increase in markers of proliferation and chromatin modifiers, global changes in DNA accessibility, and an acquisition of liver stem and progenitor cell markers. Functionally, short-term expression of 4F enhances liver regenerative capacity through topoisomerase2-mediated partial reprogramming. Our results reveal that liver-specific 4F expression in vivo induces cellular plasticity and counteracts liver failure, suggesting that partial reprogramming may represent an avenue for enhancing tissue regeneration.


Asunto(s)
Reprogramación Celular , Hígado , Animales , Desdiferenciación Celular , Hepatocitos/metabolismo , Hígado/metabolismo , Regeneración Hepática , Mamíferos , Ratones
8.
Nat Aging ; 2(3): 243-253, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-37118377

RESUMEN

Partial reprogramming by expression of reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) for short periods of time restores a youthful epigenetic signature to aging cells and extends the life span of a premature aging mouse model. However, the effects of longer-term partial reprogramming in physiologically aging wild-type mice are unknown. Here, we performed various long-term partial reprogramming regimens, including different onset timings, during physiological aging. Long-term partial reprogramming lead to rejuvenating effects in different tissues, such as the kidney and skin, and at the organismal level; duration of the treatment determined the extent of the beneficial effects. The rejuvenating effects were associated with a reversion of the epigenetic clock and metabolic and transcriptomic changes, including reduced expression of genes involved in the inflammation, senescence and stress response pathways. Overall, our observations indicate that partial reprogramming protocols can be designed to be safe and effective in preventing age-related physiological changes. We further conclude that longer-term partial reprogramming regimens are more effective in delaying aging phenotypes than short-term reprogramming.


Asunto(s)
Envejecimiento Prematuro , Reprogramación Celular , Animales , Ratones , Reprogramación Celular/genética , Envejecimiento/genética , Senescencia Celular , Envejecimiento Prematuro/genética , Modelos Animales de Enfermedad
9.
Gastroenterology ; 160(6): 2089-2102.e12, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33577875

RESUMEN

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is an effective therapy for recurrent Clostridioides difficile infection (rCDI). However, the overall mechanisms underlying FMT success await comprehensive elucidation, and the safety of FMT has recently become a serious concern because of the occurrence of drug-resistant bacteremia transmitted by FMT. We investigated whether functional restoration of the bacteriomes and viromes by FMT could be an indicator of successful FMT. METHODS: The human intestinal bacteriomes and viromes from 9 patients with rCDI who had undergone successful FMT and their donors were analyzed. Prophage-based and CRISPR spacer-based host bacteria-phage associations in samples from recipients before and after FMT and in donor samples were examined. The gene functions of intestinal microorganisms affected by FMT were evaluated. RESULTS: Metagenomic sequencing of both the viromes and bacteriomes revealed that FMT does change the characteristics of intestinal bacteriomes and viromes in recipients after FMT compared with those before FMT. In particular, many Proteobacteria, the fecal abundance of which was high before FMT, were eliminated, and the proportion of Microviridae increased in recipients. Most temperate phages also behaved in parallel with the host bacteria that were altered by FMT. Furthermore, the identification of bacterial and viral gene functions before and after FMT revealed that some distinctive pathways, including fluorobenzoate degradation and secondary bile acid biosynthesis, were significantly represented. CONCLUSIONS: The coordinated action of phages and their host bacteria restored the recipients' intestinal flora. These findings show that the restoration of intestinal microflora functions reflects the success of FMT.


Asunto(s)
Enterocolitis Seudomembranosa/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Viroma , Adulto , Anciano , Bacteriófagos , Clostridioides difficile , Enterocolitis Seudomembranosa/microbiología , Heces/microbiología , Femenino , Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/virología , Humanos , Masculino , Metagenómica , Microviridae , Persona de Mediana Edad , Proteobacteria , Viroma/genética
10.
Cell Res ; 29(10): 804-819, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31444470

RESUMEN

In vivo genome editing represents a powerful strategy for both understanding basic biology and treating inherited diseases. However, it remains a challenge to develop universal and efficient in vivo genome-editing tools for tissues that comprise diverse cell types in either a dividing or non-dividing state. Here, we describe a versatile in vivo gene knock-in methodology that enables the targeting of a broad range of mutations and cell types through the insertion of a minigene at an intron of the target gene locus using an intracellularly linearized single homology arm donor. As a proof-of-concept, we focused on a mouse model of premature-aging caused by a dominant point mutation, which is difficult to repair using existing in vivo genome-editing tools. Systemic treatment using our new method ameliorated aging-associated phenotypes and extended animal lifespan, thus highlighting the potential of this methodology for a broad range of in vivo genome-editing applications.


Asunto(s)
Edición Génica/métodos , Animales , Sistemas CRISPR-Cas/genética , Reparación del ADN , Dependovirus/genética , Factor de Transcripción GATA3/genética , Técnicas de Sustitución del Gen , Terapia Genética/métodos , Vectores Genéticos/metabolismo , Células Madre Embrionarias Humanas , Humanos , Intrones , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neuronas/citología , Neuronas/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Ratas , Tubulina (Proteína)/genética
11.
Nat Med ; 25(3): 419-422, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30778240

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare lethal genetic disorder characterized by symptoms reminiscent of accelerated aging. The major underlying genetic cause is a substitution mutation in the gene coding for lamin A, causing the production of a toxic isoform called progerin. Here we show that reduction of lamin A/progerin by a single-dose systemic administration of adeno-associated virus-delivered CRISPR-Cas9 components suppresses HGPS in a mouse model.


Asunto(s)
Sistemas CRISPR-Cas , Terapia Genética/métodos , Lamina Tipo A/genética , Longevidad , Progeria/genética , Animales , Modelos Animales de Enfermedad , Lamina Tipo A/metabolismo , Ratones , Mutación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
12.
Nature ; 561(7722): 243-247, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30185909

RESUMEN

Large cutaneous ulcers are, in severe cases, life threatening1,2. As the global population ages, non-healing ulcers are becoming increasingly common1,2. Treatment currently requires the transplantation of pre-existing epithelial components, such as skin grafts, or therapy using cultured cells2. Here we develop alternative supplies of epidermal coverage for the treatment of these kinds of wounds. We generated expandable epithelial tissues using in vivo reprogramming of wound-resident mesenchymal cells. Transduction of four transcription factors that specify the skin-cell lineage enabled efficient and rapid de novo epithelialization from the surface of cutaneous ulcers in mice. Our findings may provide a new therapeutic avenue for treating skin wounds and could be extended to other disease situations in which tissue homeostasis and repair are impaired.


Asunto(s)
Reprogramación Celular , Células Epiteliales/citología , Úlcera Cutánea/patología , Piel/citología , Heridas y Lesiones/patología , Animales , Linaje de la Célula , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Medicina Regenerativa , Piel/patología , Úlcera Cutánea/terapia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cicatrización de Heridas , Heridas y Lesiones/terapia
13.
Nature ; 540(7631): 144-149, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27851729

RESUMEN

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Marcación de Gen/métodos , Genoma/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Animales , División Celular , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Terapia Genética/métodos , Neuronas/citología , Neuronas/metabolismo , Ratas , Homología de Secuencia
14.
PLoS One ; 11(8): e0160882, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27500926

RESUMEN

Tumor-targeting Salmonella enterica serovar Typhimurium A1-R (Salmonella A1-R) had strong efficacy on a melanoma patient-derived orthotopic xenograft (PDOX) nude-mouse model. GFP-expressing Salmonella A1-R highly and selectively colonized the PDOX melanoma and significantly suppressed tumor growth (p = 0.021). The combination of Salmonella A1-R and cisplatinum (CDDP), both at low-dose, also significantly suppressed the growth of the melanoma PDOX (P = 0.001). Salmonella A1-R has future clinical potential for combination chemotherapy with CDDP of melanoma, a highly-recalcitrant cancer.


Asunto(s)
Antineoplásicos/farmacología , Modelos Animales de Enfermedad , Melanoma/terapia , Infecciones por Salmonella/microbiología , Salmonella typhimurium/crecimiento & desarrollo , Animales , Terapia Combinada , Humanos , Melanoma/microbiología , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Sci Rep ; 5: 17860, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26658478

RESUMEN

Extracellular matrix (ECM) is known to play several important roles in vascular development, although the molecular mechanisms behind these remain largely unknown. RECK, a tumor suppressor downregulated in a wide variety of cancers, encodes a membrane-anchored matrix-metalloproteinase-regulator. Mice lacking functional Reck die in utero, demonstrating its importance for mammalian embryogenesis; however, the underlying causes of mid-gestation lethality remain unclear. Using Reck conditional knockout mice, we have now demonstrated that the lack of Reck in vascular mural cells is largely responsible for mid-gestation lethality. Experiments using cultured aortic explants further revealed that Reck is essential for at least two events in sprouting angiogenesis; (1) correct association of mural and endothelial tip cells to the microvessels and (2) maintenance of fibronectin matrix surrounding the vessels. These findings demonstrate the importance of appropriate cell-cell interactions and ECM maintenance for angiogenesis and the involvement of Reck as a critical regulator of these events.


Asunto(s)
Vasos Sanguíneos/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Neovascularización Fisiológica/genética , Alelos , Animales , Matriz Extracelular/metabolismo , Femenino , Fibronectinas/metabolismo , Proteínas Ligadas a GPI/deficiencia , Silenciador del Gen , Ratones , Ratones Noqueados , Ratones Transgénicos , Microvasos/metabolismo , Mutación , Especificidad de Órganos/genética
17.
PLoS One ; 10(8): e0134324, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26237416

RESUMEN

A patient-derived nude-mouse model of soft-tissue sarcoma has been established and treated in the following groups: (1) untreated controls; (2) gemcitabine (GEM) (80 mg/kg, ip, weekly, 3 weeks); (3) Pazopanib (100 mg/kg, orally, daily, 3 weeks) and (4) Salmonella typhimurium A1-R (5 × 10(7) CFU/body, ip, weekly, 3 weeks). The sarcoma was resistant to GEM (p = 0.879). Pazopanib tended to reduce the tumor volume compared to the untreated mice, but there was no significant difference (p = 0.115). S. typhimurium A1-R significantly inhibited tumor growth compared to the untreated mice (p = 0.001). S. typhimurium A1-R was the only effective treatment for the soft-tissue sarcoma nude mouse model among all treatments including a newly approved multiple tyrosine kinase inhibitor; Pazopanib. These results suggest tumor-targeting S. typhimurium A1-R is a promising treatment for chemo-resistant soft-tissue sarcoma.


Asunto(s)
Desoxicitidina/análogos & derivados , Salmonella typhimurium , Sarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Indazoles , Ratones , Ratones Desnudos , Pirimidinas/uso terapéutico , Infecciones por Salmonella/patología , Sarcoma/patología , Neoplasias de los Tejidos Blandos/patología , Sulfonamidas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
18.
PLoS One ; 10(6): e0120358, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26047477

RESUMEN

We have previously developed mouse models of HER-2-positive cervical cancer. Tumors in nude mice had histological structures similar to the original tumor and were stained by anti-HER-2 antibody in the same pattern as the patient's cancer. We have also previously developed tumor-targeting Salmonella typhimurium A1-R and have demonstrated its efficacy against patient-derived tumor mouse models, both alone and in combination. In the current study, we determined the efficacy of S. typhimurium A1-R in combination with trastuzumab on a patient-cancer nude-mouse model of HER-2 positive cervical cancer. Mice were randomized to 5 groups and treated as follows: (1) no treatment; (2) carboplatinum (30 mg/kg, ip, weekly, 5 weeks); (3) trastuzumab (20 mg/kg, ip, weekly, 5 weeks); (4) S. typhimurium A1-R (5 × 107 CFU/body, ip, weekly, 5 weeks); (5) S. typhimurium A1-R (5 × 107 CFU/body, ip, weekly, 5 weeks) + trastuzumab (20 mg/kg, ip, weekly, 5 weeks). All regimens had significant efficacy compared to the untreated mice. The relative tumor volume of S. typhimurium A1-R + trastuzumab-treated mice was smaller compared to trastuzumab alone (p = 0.007) and S. typhimurium A1-R alone (p = 0.039). No significant body weight loss was found compared to the no treatment group except for carboplatinum-treated mice (p = 0.021). Upon histological examination, viable tumor cells were not detected, and replaced by stromal cells in the tumors treated with S. typhimurium A1-R + trastuzumab. The results of the present study suggest that S. typhimurium A1-R and trastuzumab in combination are highly effective against HER-2-expressing cervical cancer.


Asunto(s)
Receptor ErbB-2/metabolismo , Salmonella typhimurium/fisiología , Trastuzumab/uso terapéutico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Trasplante Heterólogo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
19.
Oncotarget ; 6(15): 13133-48, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26033451

RESUMEN

Fluorescence-guided surgery (FGS) of cancer is an area of intense interest. However, FGS of cancer has not yet been shown to be curative due to residual microscopic disease. Human fibrosarcoma HT1080 expressing red fluorescent protein (RFP) was implanted orthotopically in the quadriceps femoris muscle of nude mice. The tumor-bearing mice were injected with high and low-dose telomerase-dependent, green fluorescent protein (GFP)-containing adenovirus OBP-401, which labeled the tumor with GFP. Fluorescence-guided surgery (FGS) or bright light surgery (BLS) was then performed. OBP-401 could label soft-tissue sarcoma (STS) with GFP in situ, concordant with RFP. OBP-401-based FGS resulted in superior resection of STS in the orthotopic model of soft-tissue sarcoma, compared to BLS. High-dose administration of OBP-401 enabled FGS without residual sarcoma cells or local or metastatic recurrence, due to its dual effect of cancer-cell labeling with GFP and killing. High-dose OBP-401 based-FGS improved disease free survival (p = 0.00049) as well as preserved muscle function compared with BLS. High-dose OBP-401-based FGS could cure STS, a presently incurable disease. Since the parent virus of OBP-401, OBP-301, has been previously proven safe in a Phase I clinical trial, it is expected the OBP-401-FGS technology described in the present report should be translatable to the clinic in the near future.


Asunto(s)
Adenoviridae/genética , Sustancias Luminiscentes , Imagen Óptica/métodos , Sarcoma Experimental/cirugía , Neoplasias de los Tejidos Blandos/cirugía , Animales , Línea Celular Tumoral , Células Cultivadas , Fluorescencia , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Desnudos , Sarcoma Experimental/genética , Neoplasias de los Tejidos Blandos/genética , Cirugía Asistida por Computador , Proteína Fluorescente Roja
20.
Oncotarget ; 6(13): 11369-77, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25957417

RESUMEN

UNLABELLED: Peritoneal disseminated cancer is highly treatment resistant. We here report the efficacy of intraperitoneal (i.p.) administration of tumor-targeting Salmonella typhimurium A1-R in a nude mouse model of disseminated human ovarian cancer. The mouse model was established by intraperitoneal injection of the human ovarian cancer cell line SKOV3-GFP. Seven days after implantation, mice were treated with S. typhimurium A1-R via intravenous (i.v.) or i.p. administration at the same dose, 5 × 10(7) CFU, once per week. Both i.v. and i.p. treatments effected prolonged survival compared with the untreated control group (P=0.025 and P<0.001, respectively). However, i.p. treatment was less toxic than i.v. TREATMENT: Tumor-specific targeting of S. typhimurium A1-R was confirmed with bacterial culture from tumors and various organs and tumor or organ colony formation after i.v. or i.p. injection. Selective tumor targeting was most effective with i.p. administration. The results of the present study show S. typhimurium A1-R has promising clinical potential for disseminated ovarian cancer, especially via i.p. administration.


Asunto(s)
Terapia Biológica/métodos , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/terapia , Salmonella typhimurium/patogenicidad , Animales , Carga Bacteriana , Línea Celular Tumoral , Femenino , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Ratones Desnudos , Neoplasias Ováricas/microbiología , Neoplasias Ováricas/patología , Neoplasias Peritoneales/microbiología , Neoplasias Peritoneales/patología , Salmonella typhimurium/crecimiento & desarrollo , Factores de Tiempo , Virulencia , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...