Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Heliyon ; 10(7): e28676, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38617951

RESUMEN

Non-viral gene delivery is a new therapeutic in the treating genetic disorders. The most important challenge in nonviral gene transformation is the immunogenicity of carriers. Nowadays, The immunogenicity of nanocarriers as a deliverer of nucleic acid molecules has received significant attention. In this research, hematite green nanocarriers were prepared in one step with rosemary extract. Synthetic nanocarriers were investigated by using XRD (X-ray diffraction analysis), FESEM-EDX (field emission scanning electron microscopy with energy dispersive X-Ray spectroscopy), HR-TEM (high-resolution transmission electron microscopy), VSM (value stream mapping), TGA- DTG (thermal gravimetric analysis-differential thermal analysis), FT-IR (fourier-transform infrared spectroscopy), BET (brunauer-emmett-teller) and BJH (barrett-joyner-halenda) analyses. The cytotoxicity of synthetic nanocarriers was evaluated on HEK-293Tcell lines at concentration of 1-500 µg/ml using MTT method. Finally, targeted transfection of GFP plasmid using green porous particles was performed using an external magnetic field. Biogenic hematite nanoparticles with hexagonal crystal structures have a 3D pile flower-like morphology. The existence of rosemary phytochemicals in the construction of nanoparticles has caused minimal toxicity and high biocompatibility of nanocarriers. Also, TGA studies confirmed the stability of bionic nanoparticles. Superparamagnetic green nanocarriers at concentrations above 500 µg/ml is not toxic to HEK293T cells. The delivery efficiency of the plasmid was optimal at an N/P ratio of 3. Therefore, the porous α-Fe2O3 green nanocarriers are non-viral and safe carriers with potential applications in gene therapy.

2.
Biomedicines ; 11(9)2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37761005

RESUMEN

Human epidermal growth factor receptor 2 (HER2) is overexpressed in numerous cancer cell types. Therapeutic antibodies and chimeric antigen receptors (CARs) against HER2 were developed to treat human tumors. The major limitation of anti-HER2 CAR-T lymphocyte therapy is attributable to the low HER2 expression in a wide range of normal tissues. Thus, side effects are caused by CAR lymphocyte "on-target off-tumor" reactions. We aimed to develop safer HER2-targeting CAR-based therapy. CAR constructs against HER2 tumor-associated antigen (TAA) for transient expression were delivered into target T and natural killer (NK) cells by an effective and safe non-viral transfection method via nucleofection, excluding the risk of mutations associated with viral transduction. Different in vitro end-point and real-time assays of the CAR lymphocyte antitumor cytotoxicity and in vivo human HER2-positive tumor xenograft mice model proved potent cytotoxic activity of the generated CAR-T-NK cells. Our data suggest transient expression of anti-HER2 CARs in plasmid vectors by human lymphocytes as a safer treatment for HER2-positive human cancers. We also conducted preliminary investigations to elucidate if fucosylated chondroitin sulfate may be used as a possible agent to decrease excessive cytokine production without negative impact on the CAR lymphocyte antitumor effect.

3.
Adv Mater ; 35(44): e2304122, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37434421

RESUMEN

Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.


Asunto(s)
Linfoma , Receptores de Antígenos de Linfocitos T , Humanos , Linfocitos T , Transfección , Electroporación , Linfoma/metabolismo
4.
3 Biotech ; 13(3): 83, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36798854

RESUMEN

The therapeutic potential of adipose tissue-derived mesenchymal stem cells (ADMSCs) is well studied for use in non-healing wounds. However, concerns on the transplantable cell number requirement, cell expansion, cell viability, retained cell multipotency and the limited cell implantation time for efficient impact hinders cell therapy. Recent literature is much inclined to the superiority of the ADMSCs' secretome, pre-dominating its paracrine-mediated therapeutic impact. In this context, the possibility of attaining accelerated wound angiogenesis through non-viral mediated enrichment of the ADMSCs secretome with pro-angiogenic growth factors (AGF) seems promising. Accordingly, this study aimed to explore the effect of AGF-enriched ADMSCs secretome for accelerating wound angiogenesis and repair in acute large area full thickness excision rabbit wound model, as adopted from Salgado et al. (Chir Buchar Rom 108:706-710, 1990). Using sub-dermal single-dose injections along the margin of the dorsal wound, native ADMSCs secretome, AGF-enriched ADMSC secretome, allogenic rabbit ADMSCs and a combination of AGF-enriched ADMSC secretome with allogenic rabbit ADMSCs were transplanted independently. Twenty-eight days (28 days) post-transplantation, histopathological analysis was performed to assess the effect. Hematoxylin and eosin (H&E) staining showed enhanced epithelization, notable granulation tissue and collagen fiber deposition in AGF-enriched secretome transplanted groups. This was confirmed by elevated CD31 detection, faster wound closure time and collagen organization. The use of single-dose AGF-enriched ADMSCs' secretome for therapeutic angiogenesis and wound repair seems to be a promising cell-free therapeutic option. Further investigations using multiple doses on larger animal groups remains to be explored in order to ascertain the comparative potential of AGF-enriched ADMSCs' secretome.

5.
Int J Mol Sci ; 23(21)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36361771

RESUMEN

More and more patients suffer from multifactorial neurodegenerative diseases, such as age-related macular degeneration (AMD). However, their pathological mechanisms are still poorly understood, which complicates the development of effective therapies. To improve treatment of multifactorial diseases, cell-based gene therapy can be used to increase the expression of therapeutic factors. To date, there is no approved therapy for dry AMD, including late-stage geographic atrophy. We present a treatment option for dry AMD that transfers the brain-derived neurotrophic factor (BDNF) gene into retinal pigment epithelial (RPE) cells by electroporation using the plasmid-based Sleeping Beauty (SB) transposon system. ARPE-19 cells and primary human RPE cells were co-transfected with two plasmids encoding the SB100X transposase and the transposon carrying a BDNF transcription cassette. We demonstrated efficient expression and secretion of BDNF in both RPE cell types, which were further increased in ARPE-19 cell cultures exposed to hydrogen peroxide. BDNF-transfected cells exhibited lower apoptosis rates and stimulated neurite outgrowth in human SH-SY5Y cells. This study is an important step in the development of a cell-based BDNF gene therapy that could be applied as an advanced therapy medicinal product to treat dry AMD or other degenerative retinal diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Neuroblastoma , Humanos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Neuroblastoma/metabolismo , Terapia Genética , Células Epiteliales/metabolismo , Pigmentos Retinianos/metabolismo
6.
Front Physiol ; 13: 819767, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35283767

RESUMEN

In the era of the advanced nanomaterials, use of nanoparticles has been highlighted in biomedical research. However, the demonstration of DNA plasmid delivery with nanoparticles for in vivo gene delivery experiments must be carefully tested due to many possible issues, including toxicity. The purpose of the current study was to deliver a Notch Intracellular Domain (NICD)-encoded plasmid via poly(lactic-co-glycolic acid) (PLGA) nanoparticles and to investigate the toxic environmental side effects for an in vivo experiment. In addition, we demonstrated the target delivery to the endothelium, including the endocardial layer, which is challenging to manipulate gene expression for cardiac functions due to the beating heart and rapid blood pumping. For this study, we used a zebrafish animal model and exposed it to nanoparticles at varying concentrations to observe for specific malformations over time for toxic effects of PLGA nanoparticles as a delivery vehicle. Our nanoparticles caused significantly less malformations than the positive control, ZnO nanoparticles. Additionally, the NICD plasmid was successfully delivered by PLGA nanoparticles and significantly increased Notch signaling related genes. Furthermore, our image based deep-learning analysis approach evaluated that the antibody conjugated nanoparticles were successfully bound to the endocardium to overexpress Notch related genes and improve cardiac function such as ejection fraction, fractional shortening, and cardiac output. This research demonstrates that PLGA nanoparticle-mediated target delivery to upregulate Notch related genes which can be a potential therapeutic approach with minimum toxic effects.

8.
Front Cardiovasc Med ; 8: 707897, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34651022

RESUMEN

Notch signaling is a highly conserved signaling system that is required for embryonic development and regeneration of organs. When the signal is lost, maldevelopment occurs and leads to a lethal state. Delivering exogenous genetic materials encoding Notch into cells can reestablish downstream signaling and rescue cellular functions. In this study, we utilized the negatively charged and FDA approved polymer poly(lactic-co-glycolic acid) to encapsulate Notch Intracellular Domain-containing plasmid in nanoparticles. We show that primary human umbilical vein endothelial cells (HUVECs) readily uptake the nanoparticles with and without specific antibody targets. We demonstrated that our nanoparticles are non-toxic, stable over time, and compatible with blood. We further demonstrated that HUVECs could be successfully transfected with these nanoparticles in static and dynamic environments. Lastly, we elucidated that these nanoparticles could upregulate the downstream genes of Notch signaling, indicating that the payload was viable and successfully altered the genetic downstream effects.

9.
Biotechnol Prog ; 37(4): e3152, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33774920

RESUMEN

Cultured fibroblast cells, especially dermal cells, are used for various types of scientific research, particularly within the medical field. Desirable features of the cells include their ease of isolation, rapid cellular growth, and high degree of robustness. Currently, fibroblasts are mainly used to obtain pluripotent cells via a reprogramming process. Dermal fibroblasts, are particularly useful for gene therapies used for promoting wound healing or minimizing skin aging. In recent years, fibroblast transfection efficiencies have significantly improved. In order to introduce molecules (most often DNA or RNA) into cells, viral-based systems (transduction) or non-viral methods (transfection) that include physical/mechanical processes or lipid reagents may be used. In this article, we describe critical points that should be considered when selecting a method for transfecting fibroblasts. The most effective methods used for the transfection of fibroblasts include both viral-based and non-viral nucleofection systems. These methods result in a high level of transgene expression and are superior in terms of transfection efficacy and viability.


Asunto(s)
Fibroblastos , Terapia Genética , Línea Celular , Células Cultivadas , Fibroblastos/metabolismo , Transfección
10.
J Biomed Mater Res B Appl Biomater ; 108(7): 2807-2819, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32243682

RESUMEN

Cell-based skin substitute generation has seen considerable development. Combining synthetic scaffolds with biomimetic fibrin does direct both exogenous and endogenous stem cell differentiation, addressing needs for reliable tissue engineering. However, lack of immediate vasculature within implantable grafts remains critical for its sustenance and integration. Multipotency, high proliferation potential, ability to release multiple growth factors (GFs), and autologous availability highlight the use of human adipose derived mesenchymal stem cells (hADMSCs) in tissue-engineered dermal grafts (TEDG) construction. However, hADMSCs' insufficiency to independently establish angiogenesis within tissue constructs demands improvement of stem cell application for dermal graft survival. Approaches to harness microenvironmentally sensitive paracrine interactions could improve the angiogenic efficiency of hADMSCs within TEDG. This study conceptualized a fibrin-based niche, to direct hADMSCs toward a nonfibrotic fibroblast commitment and incorporation of bioengineered hADMSCs, specifically releasing potent angiogenic factors within TEDG. Coexistence of tuned fibroblast and endothelial lineage committed cells contributed to well-regulated extracellular matrix formation and prevascularization. Adequate cell proliferation; sustained transient release of angiogenic GFs till 20 days; directed dermal, endothelial, fibroblast, and vascular smooth muscle cell differentiation; and favored elastin and collagen deposition were achieved in vitro. In conclusion, specific niche composition and employment of bioengineered hADMSCs favor implantable TEDG construction.


Asunto(s)
Tejido Adiposo/metabolismo , Diferenciación Celular , Dermis , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Piel Artificial , Ingeniería de Tejidos , Matriz Extracelular/metabolismo , Humanos
11.
J Gene Med ; 20(10-11): e3054, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30172246

RESUMEN

BACKGROUND: Cervical cancer is the third most common cause of cancer in women. The 5-year survival rate in oropharyngeal squamous cell carcinomas is approximately 50% and this rate has not improved in recent decades. These cancers are accessible to direct intervention. We examined the ability of a highly efficient non-viral vector, TransfeX (ATCC, Manassas, VA, USA), to deliver the suicide gene HSV-tk to cervical, oral and pharyngeal cancer cells and to induce cytotoxicity following the administration of the prodrug, ganciclovir. METHODS: HeLa cervical carcinoma, HSC-3 and H357 oral squamous cell carcinoma and FaDu pharyngeal carcinoma cells were transfected with cytomegalovirus (CMV)- or enhanced episomal vector (EEV)-driven HSV-tk plasmids and treated with ganciclovir for 24-120 h. Cell viability was assessed by Alamar blue. RESULTS: The viability of HeLa cells was reduced to only 30-40%, despite the very high levels of transgene expression. By contrast, the viability of HSC-3 cells was reduced to 10%, although transgene expression was 18-fold lower than that in HeLa cells. An approximately five-fold higher transgene expression was obtained with the EEV-plasmid than from the CMV-plasmid. Nevertheless, HeLa cell viability after suicide gene + ganciclovir treatment was reduced by only 35% compared to 70% with the CMV-plasmid. For HSC-3 cells, the reduction was 40% for the EEV- and 80% for the CMV-plasmid. The lower efficiency of transfection with the EEV-plasmid may explain the lower cytotoxicity. CONCLUSIONS: TransfeX-mediated gene delivery to cervical, pharyngeal and oral cancer cells may be used for suicide gene therapy. The levels of transgene expression, however, do not translate directly to cytotoxicity.


Asunto(s)
Citomegalovirus/genética , Técnicas de Transferencia de Gen , Genes Transgénicos Suicidas/genética , Plásmidos/genética , Antivirales/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Ganciclovir/farmacología , Terapia Genética/métodos , Vectores Genéticos/genética , Células HeLa , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Neoplasias Faríngeas/genética , Neoplasias Faríngeas/patología , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
12.
Biotechnol Adv ; 36(7): 1855-1869, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30012541

RESUMEN

Current protocols for chondrocyte expansion and chondrogenic differentiation of stem cells fail to reduce phenotypic loss and to mitigate hypertrophic tendency. To this end, cell genetic manipulation is gaining pace as a means of generating cells with stable chondrocyte phenotype. Herein, we provide an overview of candidate genes that either induce cartilage regeneration or inhibit cartilage degeneration. We further discuss in vitro, ex vivo and in vivo viral transduction and non-viral transfection strategies for targeted cells (chondrocytes, mesenchymal stem cells, induced pluripotent stem cells and synovial cells), along with the most representative results obtained in pre-clinical models and in clinical trials. We highlight current challenges and associated risks that slowdown clinical acceptance and commercialisation of gene transfer technologies.


Asunto(s)
Artritis Reumatoide/terapia , Cartílago/fisiología , Condrogénesis/genética , Ingeniería Genética , Osteoartritis/terapia , Regeneración/genética , Diferenciación Celular/genética , Condrocitos/fisiología , Terapia Genética , Humanos , Células Madre Mesenquimatosas/fisiología , Fenotipo , Células Madre/fisiología
13.
Front Immunol ; 8: 536, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28553285

RESUMEN

Mesenchymal stem cells (MSCs) are currently exploited as gene delivery systems for transient in situ expression of cancer therapeutics. As an alternative to the prevailing viral expression, we here describe a murine MSC line stably expressing a therapeutic protein for up to 42 passages, yet fully maintaining MSC features. Because of superior antitumoral activity of hexavalent TNF-related apoptosis-inducing ligand (TRAIL) formats and the advantage of a tumor-targeted action, we choose expression of a dimeric EGFR-specific diabody single-chain TRAIL (Db-scTRAIL) as a model. The bioactivity of Db-scTRAIL produced from an isolated clone (MSC.TRAIL) was revealed from cell death induction in Colo205 cells treated with either culture supernatants from or cocultured with MSC.TRAIL. In vivo, therapeutic activity of MSC.TRAIL was shown upon peritumoral injection in a Colo205 xenograft tumor model. Best antitumor activity in vitro and in vivo was observed upon combined treatment of MSC.TRAIL with bortezomib. Importantly, in vivo combination treatment did not cause apparent hepatotoxicity, weight loss, or behavioral changes. The development of well characterized stocks of stable drug-producing human MSC lines has the potential to establish standardized protocols of cell-based therapy broadly applicable in cancer treatment.

14.
J Neurol Sci ; 375: 137-145, 2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28320116

RESUMEN

Neurotrophin 3 (NT-3) is an important factor for promoting prenatal neural development, as well as regeneration, axogenesis and plasticity in postnatal life. Therapy with NT-3 was reported to improve the condition of patients suffering from degenerative diseases and traumatic injuries, however, the disadvantage of NT-3 protein delivery is its short half-life, thus our alternative approach is the use of NT-3 gene therapy. In this study, the bone marrow stromal cells (BMSCs) were isolated from adult rats, cultured for 4 passages and transfected with either pEGFP-N1 or a constructed vector containing murine proNT-3 (pSecTag2/HygroB-murine proNT-3) using Lipofectamine 2000 followed by Hygromycin B (200mg/kg). The transfection efficiency of the transiently transfected BMSCs was evaluated using the green fluorescence protein containing vector (pEGFP-N1). A quantitative evaluation of the NT-3 expression of mRNA using real time qRT-PCR shows that there was double fold increase in NT-3 gene expression compared with non-transfected BMSCs, also, the culture supernatant yielded double fold increase in NT-3 using ELISA technique, the data were supported by immunoblotting technique. This suggests that the use of this transfection technique can be useful for gene therapy in different neurological disorders with neurodegenerative or traumatic origins.


Asunto(s)
Células de la Médula Ósea/metabolismo , Neurotrofina 3/genética , Transfección , Animales , Antígenos CD/metabolismo , Células Cultivadas , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Neurotrofina 3/metabolismo , ARN Mensajero/metabolismo , Ratas
15.
Springerplus ; 5(1): 1521, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27652094

RESUMEN

BACKGROUND: CRISPR-Cas9 genome editing and labeling has emerged as an important tool in biologic research, particularly in regards to potential transgenic and gene therapy applications. Delivery of CRISPR-Cas9 plasmids to target cells is typically done by non-viral methods (chemical, physical, and/or electrical), which are limited by low transfection efficiencies or with viral vectors, which are limited by safety and restricted volume size. In this work, a non-viral transfection technology, named lance array nanoinjection (LAN), utilizes a microfabricated silicon chip to physically and electrically deliver genetic material to large numbers of target cells. To demonstrate its utility, we used the CRISPR-Cas9 system to edit the genome of isogenic cells. Two variables related to the LAN process were tested which include the magnitude of current used during plasmid attraction to the silicon lance array (1.5, 4.5, or 6.0 mA) and the number of times cells were injected (one or three times). RESULTS: Results indicate that most successful genome editing occurred after injecting three times at a current control setting of 4.5 mA, reaching a median level of 93.77 % modification. Furthermore, we found that genome editing using LAN follows a non-linear injection-dose response, meaning samples injected three times had modification rates as high as nearly 12 times analogously treated single injected samples. CONCLUSIONS: These findings demonstrate the LAN's ability to deliver genetic material to cells and indicate that successful alteration of the genome is influenced by a serial injection method as well as the electrical current settings.

16.
J Neurol Sci ; 339(1-2): 81-6, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24518204

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is one of the most important proteins playing a pivotal role in growing and repairing of the nervous system. GDNF therapy is one of the suggested options in the treatment of neurodegenerative diseases. Limitations in the viral gene delivery and its side effects after therapy have encouraged us to use a non-viral method one for this purpose. We transfected rat bone marrow stromal cells (BMSCs) in ex vivo conditions using Lipofectamine 2000 reagent with pEGFP-C1 and a constructed vector carrying the human proGDNF (pSecTag2/HygroB-human proGDNF), transiently and stably, respectively. The rate of transient transfection of rat BMSCs was eight percent and transfected rat BMSCs with pSecTag2/HygroB-human proGDNF stabilized by adding Hygromycin B in cell culture medium at 200 µg/ml. Semi-quantitative data analysis from Western-blot technique showed that stable transfected cells secrete GDNF at higher level in comparison with control cells (6.530 fold in the supernatant). The present study supports the utility of liposome-mediated transfection for overexpressing human GDNF in rat BMSCs. For this purpose and in order to get more yield of human GDNF secretion from the stable transfected rat BMSCs, we used a vector containing another signal sequence instead of its own pre-segment of proGDNF protein. This is the first report in this regard and the data presented will be potentially useful for human gene transfer therapies in a variety of neurodegenerative diseases.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Células Madre Mesenquimatosas/fisiología , Animales , Células Cultivadas , Vectores Genéticos/biosíntesis , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Humanos , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA