Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Cell Mol Med ; 28(17): e70056, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39245805

RESUMO

Disruptions in normal development and the emergence of health conditions often result from the malfunction of vital genes in the human body. Decades of scientific research have focused on techniques to modify or substitute defective genes with healthy alternatives, marking a new era in disease treatment, prevention and cure. Recent strides in science and technology have reshaped our understanding of disorders, medication development and treatment recommendations, with human gene and cell therapy at the forefront of this transformative shift. Its primary objective is the modification of genes or adjustment of cell behaviour for therapeutic purposes. In this review, we focus on the latest advances in gene and cell therapy for treating human genetic diseases, with a particular emphasis on FDA and EMA-approved therapies and the evolving landscape of genome editing. We examine the current state of innovative gene editing technologies, particularly the CRISPR-Cas systems. As we explore the progress, ethical considerations and prospects of these innovations, we gain insight into their potential to revolutionize the treatment of genetic diseases, along with a discussion of the challenges associated with their regulatory pathways. This review traces the origins and evolution of these therapies, from conceptual ideas to practical clinical applications, marking a significant milestone in the field of medical science.


Assuntos
Sistemas CRISPR-Cas , Terapia Baseada em Transplante de Células e Tecidos , Edição de Genes , Doenças Genéticas Inatas , Terapia Genética , Humanos , Terapia Genética/métodos , Doenças Genéticas Inatas/terapia , Doenças Genéticas Inatas/genética , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Edição de Genes/métodos , Animais
2.
Eur J Neurosci ; 59(8): 1993-2015, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38382910

RESUMO

A common pathological hallmark of neurodegenerative disorders is neuronal cell death, accompanied by neuroinflammation and oxidative stress. The vasoactive intestinal peptide (VIP) is a pleiotropic peptide that combines neuroprotective and immunomodulatory actions. The gene therapy field shows long-term promise for treating a wide range of neurodegenerative diseases (ND). In this study, we aimed to investigate the in vitro efficacy of transduction of microglia using lentiviral gene therapy vectors encoding VIP (LentiVIP). Additionally, we tested the protective effects of the secretome derived from LentiVIP-infected "immortalized human" microglia HMC3 cells, and cells treated with Synthetic VIP (SynVIP), against toxin-induced neurodegeneration. First, LentiVIP, which stably expresses VIP, was generated and purified. VIP secretion in microglial conditioned media (MG CM) for LentiVIP-infected HMC3 microglia cells was confirmed. Microglia cells were activated with lipopolysaccharide, and groups were formed as follows: 1) Control, 2) SynVIP-treated, or 3) LentiVIP-transduced. These MG CM were applied on an in vitro neurodegenerative model formed by differentiated (d)-SH-SY5Y cells. Then, cell survival analysis and apoptotic nuclear staining, besides measurement of oxidative/inflammatory parameters in CM of cells were performed. Activated MG CM reduced survival rates of both control and toxin-applied (d)-SH-SY5Y cells, whereas LentiVIP-infected MG CM and SynVIP-treated ones exhibited better survival rates. These findings were supported by apoptotic nuclear evaluations of (d)-SH-SY5Y cells, alongside oxidative/inflammatory parameters in their CM. LentiVIP seems worthy of further studies for the treatment of ND because of the potential of gene therapy to treat diseases effectively with a single injection.


Assuntos
Neuroblastoma , Doenças Neurodegenerativas , Fármacos Neuroprotetores , Humanos , Peptídeo Intestinal Vasoativo/farmacologia , Peptídeo Intestinal Vasoativo/metabolismo , Microglia/metabolismo , Neuroblastoma/metabolismo , Doenças Neurodegenerativas/metabolismo , Terapia Genética , Fármacos Neuroprotetores/farmacologia
3.
Adv Exp Med Biol ; 1409: 145-159, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36289162

RESUMO

Since insulin deficiency results from pancreatic beta-cell destruction, all type 1 and most type 2 diabetes patients eventually require life-long insulin injections. Insulin gene synthesis could also be impaired due to insulin gene mutations as observed in diabetic patients with MODY 10. At this point, insulin gene therapy could be very effective to recompense insulin deficiency under these circumstances. For this reason, an HIV-based lentiviral vector carrying the insulin gene under the control of insulin promoter (LentiINS) was generated, and its therapeutic efficacy was tested in a beta-cell transplant model lacking insulin produced by CRISPR/Cas9-mediated genetically engineered pancreatic beta cells. To generate an insulin knockout beta-cell transplant animal model of diabetes, a dual gene knockout plasmid system involving CRISPR/Cas9 was transfected into a mouse pancreatic beta cell line (Min6). Fluorescence microscopy and antibiotic selection were utilized to select the insulin gene knockout clones. Transplantation of the genetically engineered pancreatic beta cells under the kidney capsule of STZ-induced diabetic rats revealed LentiINS- but not LentiLacZ-infected Ins2KO cells transiently reduced hyperglycemia similar to that of MIN6 in diabetic animals. These results suggest LentiINS has the potential to functionally restore insulin production in an insulin knockout beta-cell transplant animal model of diabetes.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Camundongos , Animais , Ratos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Insulina/genética , Insulina/metabolismo , Animais Geneticamente Modificados , Células Secretoras de Insulina/metabolismo , Modelos Animais , Transplante de Células/métodos
4.
Mol Ther ; 29(1): 149-161, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33130311

RESUMO

Autoimmune destruction of pancreatic beta cells is the characteristic feature of type 1 diabetes mellitus. Consequently, both short- and intermediate-acting insulin analogs are under development to compensate for the lack of endogenous insulin gene expression. Basal insulin is continuously released at low levels in response to hepatic glucose output, while post-prandial insulin is secreted in response to hyperglycemia following a meal. As an alternative to multiple daily injections of insulin, glucose-regulated insulin gene expression by gene therapy is under development to better endure postprandial glucose excursions. Controlled transcription and translation of proinsulin, presence of glucose-sensing machinery, prohormone convertase expression, and a regulated secretory pathway are the key features unique to pancreatic beta cells. To take advantage of these hallmarks, we generated a new lentiviral vector (LentiINS) with an insulin promoter driving expression of the proinsulin encoding cDNA to sustain pancreatic beta-cell-specific insulin gene expression. Intraperitoneal delivery of HIV-based LentiINS resulted in the lowering of fasting plasma glucose, improved glucose tolerance and prevented weight loss in streptozoticin (STZ)-induced diabetic Wistar rats. However, the combinatorial use of LentiINS and anti-inflammatory lentiviral vector (LentiVIP) gene therapy was required to increase serum insulin to a level sufficient to suppress non-fasting plasma glucose and diabetes-related inflammation.


Assuntos
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Terapia Genética , Vetores Genéticos/genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Lentivirus/genética , Animais , Biomarcadores , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Vetores Genéticos/administração & dosagem , Glucose/metabolismo , Insulina/metabolismo , Ratos , Ratos Wistar , Resultado do Tratamento
5.
Gene Ther ; 28(3-4): 130-141, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32733091

RESUMO

Type 1 diabetes (T1DM) is an autoimmune condition in which the immune system attacks and destroys insulin-producing beta cells in the pancreas leading to hyperglycemia. Vasoactive intestinal peptide (VIP) manifests insulinotropic and anti-inflammatory properties, which are useful for the treatment of diabetes. Because of its limited half-life due to DPP-4-mediated degradation, constant infusions or multiple injections are needed to observe any therapeutic benefit. Since gene therapy has the potential to treat genetic diseases, an HIV-based lentiviral vector carrying VIP gene (LentiVIP) was generated to provide a stable VIP gene expression in vivo. The therapeutic efficacy of LentiVIP was tested in a multiple low-dose STZ-induced animal model of T1DM. LentiVIP delivery into diabetic animals reduced hyperglycemia, improved glucose tolerance, and prevented weight loss. Also, a decrease in serum CRP levels, and serum oxidant capacity, but an increase in antioxidant capacity were observed in LentiVIP-treated animals. Restoration of islet cell mass was correlated with an increase in pancreatic beta-cell proliferation. These beneficial results suggest the therapeutic effect of LentiVIP is due to the repression of diabetes-induced inflammation, its insulinotropic properties, and VIP-induced beta-cell proliferation.


Assuntos
Diabetes Mellitus Tipo 1 , Peptídeo Intestinal Vasoativo , Animais , Proliferação de Células , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Terapia Genética , Inflamação/terapia , Insulina , Peptídeo Intestinal Vasoativo/genética
6.
Gene Ther ; 25(4): 269-283, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29523882

RESUMO

Type 2 diabetes mellitus (T2DM) is characterised by insulin resistance, glucose intolerance and beta cell loss leading to hyperglycemia. Vasoactive intestinal peptide (VIP) has been regarded as a novel therapeutic agent for the treatment of T2DM because of its insulinotropic and anti-inflammatory properties. Despite these beneficial properties, VIP is extremely sensitive to peptidases (DPP-4) requiring constant infusion or multiple injections to observe any therapeutic benefit. Thus, we constructed an HIV-based lentiviral vector encoding human VIP (LentiVIP) to test the therapeutic efficacy of VIP peptide in a diet-induced obesity (DIO) animal model of T2DM. VIP gene expression was shown by immunocytochemistry (ICC) and VIP peptide secretion was confirmed by ELISA both in HepG2 liver and MIN6 pancreatic beta cell lines. Functional properties of VIP were demonstrated by cAMP production assay and glucose-stimulated insulin secretion test (GSIS). Intraperitoneal (IP) delivery of LentiVIP vectors into mice significantly increased serum VIP concentrations compared to control mice. Most importantly, LentiVIP delivery in DIO animal model of T2DM resulted in improved insulin sensitivity, glucose tolerance and protection against STZ-induced diabetes in addition to reduction in serum triglyceride/cholesterol levels. Collectively, these data suggest LentiVIP delivery should be evaluated as an experimental therapeutic approach for the treatment of T2DM.


Assuntos
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/terapia , Peptídeo Intestinal Vasoativo/genética , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/prevenção & controle , Dieta Hiperlipídica , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Glucose/metabolismo , Intolerância à Glucose , Células Hep G2 , Humanos , Insulina/metabolismo , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Peptídeo Intestinal Vasoativo/administração & dosagem , Peptídeo Intestinal Vasoativo/biossíntese
7.
Expert Rev Mol Med ; 16: e7, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24666581

RESUMO

Glucagon-like peptide (GLP)-1 is an incretin hormone with several antidiabetic functions including stimulation of glucose-dependent insulin secretion, increase in insulin gene expression and beta-cell survival. Despite the initial technical difficulties and profound inefficiency of direct gene transfer into the pancreas that seriously restricted in vivo gene transfer experiments with GLP-1, recent exploitation of various routes of gene delivery and alternative means of gene transfer has permitted the detailed assessment of the therapeutic efficacy of GLP-1 in animal models of type 2 diabetes (T2DM). As a result, many clinical benefits of GLP-1 peptide/analogues observed in clinical trials involving induction of glucose tolerance, reduction of hyperglycaemia, suppression of appetite and food intake linked to weight loss have been replicated in animal models using gene therapy. Furthermore, GLP-1-centered gene therapy not only improved insulin sensitivity, but also reduced abdominal and/or hepatic fat associated with obesity-induced T2DM with drastic alterations in adipokine profiles in treated subjects. Thus, a comprehensive assessment of recent GLP-1-mediated gene therapy approaches with detailed analysis of current hurdles and resolutions, is discussed.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Peptídeo 1 Semelhante ao Glucagon/genética , Adenoviridae/genética , Animais , Dependovirus/genética , Terapia Genética , Vetores Genéticos , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Humanos , Pâncreas/metabolismo , Pâncreas/patologia , Regiões Promotoras Genéticas , Integração Viral
8.
Diabetes Metab Res Rev ; 30(5): 354-71, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24989141

RESUMO

Therapies targeting the action of incretin hormones have been under close scrutiny in recent years. The incretin effect has been defined as postprandial enhancement of insulin secretion by gut-derived factors. Likewise, incretin mimetics and incretin effect amplifiers are the two different incretin-based treatment strategies developed for the treatment of diabetes. Although, incretin mimetics produce effects very similar to those of natural incretin hormones, incretin effect amplifiers act by inhibiting dipeptidyl peptidase-4 (DPP-4) enzyme to increase plasma concentration of incretins and their biologic effects. Because glucagon-like peptide-1 (GLP-1) is an incretin hormone with various anti-diabetic actions including stimulation of glucose-induced insulin secretion, inhibition of glucagon secretion, hepatic glucose production and gastric emptying, it has been evaluated as a novel therapeutic agent for the treatment of type 2 diabetes mellitus (T2DM). GLP-1 also manifests trophic effects on pancreas such as pancreatic beta cell growth and differentiation. Because DPP-4 is the enzyme responsible for the inactivation of GLP-1, DPP-4 inhibition represents another potential strategy to increase plasma concentration of GLP-1 to enhance the incretin effect. Thus, anti-diabetic properties of these two classes of drugs have stimulated substantial clinical interest in the potential of incretin-based therapeutic agents as a means to control glucose homeostasis in T2DM patients. Despite this fact, clinical use of GLP-1 mimetics and DPP-4 inhibitors have raised substantial concerns owing to possible side effects of the treatments involving increased risk for pancreatitis, and C-cell adenoma/carcinoma. Thus, controversial issues in incretin-based therapies under development are reviewed and discussed in this manuscript.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Hipoglicemiantes/uso terapêutico , Incretinas/fisiologia , Incretinas/uso terapêutico , Animais , Diabetes Mellitus Tipo 2/fisiopatologia , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Exenatida , Polipeptídeo Inibidor Gástrico/biossíntese , Polipeptídeo Inibidor Gástrico/fisiologia , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Receptor do Peptídeo Semelhante ao Glucagon 1 , Hemoglobinas Glicadas/metabolismo , Humanos , Incretinas/efeitos adversos , Insulina/metabolismo , Insulina Glargina , Secreção de Insulina , Insulina de Ação Prolongada/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Peptídeos/uso terapêutico , Receptores de Glucagon/agonistas , Receptores de Glucagon/efeitos dos fármacos , Peçonhas/uso terapêutico
9.
Expert Rev Mol Med ; 14: e18, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23062285

RESUMO

Despite the fact that insulin injection can protect diabetic patients from developing diabetes-related complications, recent meta-analyses indicate that rapid and long-acting insulin analogues only provide a limited benefit compared with conventional insulin regarding glycemic control. As insulin deficiency is the main sequel of type-1 diabetes (T1D), transfer of the insulin gene-by-gene therapy is becoming an attractive treatment modality even though T1D is not caused by a single genetic defect. In contrast to human insulin and insulin analogues, insulin gene therapy targets to supplement patients not only with insulin but also with C-peptide. So far, insulin gene therapy has had limited success because of delayed and/or transient gene expression. Sustained insulin gene expression is now feasible using current gene-therapy vectors providing patients with basal insulin coverage, but management of postprandial hyperglycaemia is still difficult to accomplish because of the inability to properly control insulin secretion. Enteroendocrine cells of the gastrointestinal track (K cells and L cells) may be ideal targets for insulin gene therapy, but cell-targeting difficulties have limited practical implementation of insulin gene therapy for diabetes treatment. Therefore, recent gene transfer technologies developed to generate authentic beta cells through transdifferentiation are also highlighted in this review.


Assuntos
Diabetes Mellitus Tipo 1/genética , Terapia Genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Diabetes Mellitus Tipo 1/terapia , Regulação da Expressão Gênica , Humanos , Insulina/metabolismo
10.
BMC Cancer ; 12: 58, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22313795

RESUMO

BACKGROUND: Colorectal cancer is the third most common cancer and the third leading cause of cancer-related death. Bevacizumab is a humanized monoclonal antibody developed against vascular endothelial growth factor (VEGF) for the treatment of metastatic cancer. The parameters of RECIST (Response Evaluation Criteria for Solid Tumors) are not adequate to detect important treatment effects and response. Our goal was to evaluate the possibility of using sTRAIL (serum-soluble TNF-related apoptosis-inducing ligand) and VEGF as markers of treatment efficacy and prognosis in patients with metastatic colon cancer. METHODS: sTRAIL and VEGF levels were measured by ELISA in the sera of 16 bevacizumab-treated metastatic colon cancer patients and 10 presumably healthy age-matched controls. The measurements were taken before and after treatment for comparison purposes. RESULTS: Elevated levels of sTRAIL were found in seven out of 16 patients after bevacizumab treatment. Although these patients had a median survival time of 20.6 months, the remaining bevacizumab-treated patients who did not show an increase in sTRAIL had a median survival time of 9.4 months. As expected, serum VEGF levels were decreased in all patients who received bevacizumab therapy and showed no correlation between serum VEGF levels and patient survival (data not shown). CONCLUSIONS: Serum sTRAIL levels might be a useful predictor of prognosis in metastatic colon cancer, in the early evaluation stages following bevacizumab treatment.


Assuntos
Adenoma/diagnóstico , Biomarcadores Tumorais/sangue , Neoplasias do Colo/diagnóstico , Ligante Indutor de Apoptose Relacionado a TNF/sangue , Adenoma/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Bevacizumab , Neoplasias do Colo/tratamento farmacológico , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/sangue
11.
Diabetes Metab Res Rev ; 27(6): 575-83, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21584921

RESUMO

BACKGROUND: To increase the success rate in xenogeneic islet transplantation, proper assessment of graft mass is required following transplantation. For this reason, we aimed to develop a suitable fluorescence imaging system to monitor islet xenograft survival in diabetic mice. METHODS: Adenovirus vector encoding enhanced green fluorescent protein-transduced rat pancreatic islets were transplanted under the renal capsule of streptozotocin-induced diabetic mice and the fluorescence signal was quantified over time using a cooled charge-coupled device. Non-fasting blood glucose levels were recorded during the same period. Insulin release from transduced and control islets was detected via enzyme-linked immunosorbent assay. RESULTS: Adenovirus vector encoding enhanced green fluorescent protein infection did not alter the function or survival of pancreatic islets post transduction. A direct correlation was found between the number of islets (250-750) transplanted under the kidney capsule and the blood glucose recovery. CONCLUSIONS: Fluorescence imaging appears to be a useful tool for quantitative assessment of islet cell viability post transplantation and could permit earlier detection of graft rejection.


Assuntos
Sobrevivência de Enxerto/fisiologia , Proteínas de Fluorescência Verde , Transplante das Ilhotas Pancreáticas/métodos , Transplante Heterólogo , Adenoviridae/genética , Animais , Glicemia/metabolismo , Sobrevivência Celular , Diabetes Mellitus Experimental , Feminino , Fluorescência , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Wistar
12.
Hum Gene Ther ; 32(11-12): 541-562, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33858231

RESUMO

Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease (COVID-19) caused by the novel coronavirus SARS-coronavirus 2 (CoV-2). To combat the devastating spread of SARS-CoV-2, extraordinary efforts from numerous laboratories have focused on the development of effective and safe vaccines. Traditional live-attenuated or inactivated viral vaccines are not recommended for immunocompromised patients as the attenuated virus can still cause disease via phenotypic or genotypic reversion. Subunit vaccines require repeated dosing and adjuvant use to be effective, and DNA vaccines exhibit lower immune responses. mRNA vaccines can be highly unstable under physiological conditions. On the contrary, naturally antigenic viral vectors with well-characterized structure and safety profile serve as among the most effective gene carriers to provoke immune response via heterologous gene transfer. Viral vector-based vaccines induce both an effective cellular immune response and a humoral immune response owing to their natural adjuvant properties via transduction of immune cells. Consequently, viral vectored vaccines carrying the SARS-CoV-2 spike protein have recently been generated and successfully used to activate cytotoxic T cells and develop a neutralizing antibody response. Recent progress in SARS-CoV-2 vaccines, with an emphasis on gene therapy viral vector-based vaccine development, is discussed in this review.


Assuntos
Vacinas contra COVID-19/farmacologia , Vetores Genéticos , Vacinas Atenuadas/farmacologia , Vacinas Sintéticas/farmacologia , Proteínas Estruturais Virais/química , Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/química , Vetores Genéticos/genética , Humanos , Lentivirus/genética , SARS-CoV-2/genética , Vacinas de DNA/farmacologia , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Vacinas de mRNA
13.
World J Stem Cells ; 13(6): 485-502, 2021 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-34249224

RESUMO

Targeted genome editing is a continually evolving technology employing programmable nucleases to specifically change, insert, or remove a genomic sequence of interest. These advanced molecular tools include meganucleases, zinc finger nucleases, transcription activator-like effector nucleases and RNA-guided engineered nucleases (RGENs), which create double-strand breaks at specific target sites in the genome, and repair DNA either by homologous recombination in the presence of donor DNA or via the error-prone non-homologous end-joining mechanism. A recently discovered group of RGENs known as CRISPR/Cas9 gene-editing systems allowed precise genome manipulation revealing a causal association between disease genotype and phenotype, without the need for the reengineering of the specific enzyme when targeting different sequences. CRISPR/Cas9 has been successfully employed as an ex vivo gene-editing tool in embryonic stem cells and patient-derived stem cells to understand pancreatic beta-cell development and function. RNA-guided nucleases also open the way for the generation of novel animal models for diabetes and allow testing the efficiency of various therapeutic approaches in diabetes, as summarized and exemplified in this manuscript.

14.
BMC Cancer ; 10: 584, 2010 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-20977779

RESUMO

BACKGROUND: Lung cancer causes the highest rate of cancer-related deaths both in men and women. As many current treatment modalities are inadequate in increasing patient survival, new therapeutic strategies are required. TNF-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in tumor cells but not in normal cells, prompting its current evaluation in a number of clinical trials. The successful therapeutic employment of TRAIL is restricted by the fact that many tumor cells are resistant to TRAIL. The goal of the present study was to test a novel combinatorial gene therapy modality involving adenoviral delivery of TRAIL (Ad5hTRAIL) and IKK inhibition (AdIKKßKA) to overcome TRAIL resistance in lung cancer cells. METHODS: Fluorescent microscopy and flow cytometry were used to detect optimum doses of adenovirus vectors to transduce lung cancer cells. Cell viability was assessed via a live/dead cell viability assay. Luciferase assays were employed to monitor cellular NF-κB activity. Apoptosis was confirmed using Annexin V binding. RESULTS: Neither Ad5hTRAIL nor AdIKKßKA infection alone induced apoptosis in A549 lung cancer cells, but the combined use of Ad5hTRAIL and AdIKKßKA significantly increased the amount of A549 apoptosis. Luciferase assays demonstrated that both endogenous and TRAIL-induced NF-κB activity was down-regulated by AdIKKßKA expression. CONCLUSIONS: Combination treatment with Ad5hTRAIL and AdIKKßKA induced significant apoptosis of TRAIL-resistant A549 cells, suggesting that dual gene therapy strategy involving exogenous TRAIL gene expression with concurrent IKK inhibition may be a promising novel gene therapy modality to treat lung cancer.


Assuntos
Adenoviridae/genética , Regulação Neoplásica da Expressão Gênica , Quinase I-kappa B/metabolismo , Neoplasias Pulmonares/metabolismo , NF-kappa B/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Adenoviridae/metabolismo , Apoptose , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Citometria de Fluxo/métodos , Humanos , Masculino , Microscopia de Fluorescência/métodos
15.
Urol Int ; 84(4): 445-51, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20339295

RESUMO

BACKGROUND: It is not known whether androgen ablation therapy (AAT) influences TRAIL death ligand and its receptors expression of prostate cancer (PCa) cells. AIM: To investigate whether hormonal therapy alters the expression of TRAIL death ligand and TRAIL receptors in patients with advanced PCa. PATIENTS AND METHODS: 26 untreated and 20 AAT-treated advanced PCa patients were included in the study. The patients who received AAT were divided into two groups based on hormone sensitivity status. TRAIL ligand and receptor expression were determined by a conventional immunohistochemistry method. RESULTS: TRAIL death ligand and TRAIL-R2 death receptor were upregulated in PCa patients who received AAT. Hormone-refractory PCa patients exhibited lower levels of TRAIL death receptor (TRAIL-R1 and TRAIL-R2) expression compared to hormone-sensitive PCa patients. CONCLUSIONS: AAT alters TRAIL death ligand and its receptors expression in patients with PCa.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Resistencia a Medicamentos Antineoplásicos , Humanos , Imuno-Histoquímica , Masculino , Estadiamento de Neoplasias , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fatores de Tempo , Resultado do Tratamento , Turquia
16.
BMC Musculoskelet Disord ; 11: 192, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20799941

RESUMO

BACKGROUND: Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disorder. Although the pathogenesis of disease is unclear, it is well known that T cells play a major role in both development and perpetuation of RA through activating macrophages and B cells. Since the lack of TNF-Related Apoptosis Inducing Ligand (TRAIL) expression resulted in defective thymocyte apoptosis leading to an autoimmune disease, we explored evidence for alterations in TRAIL/TRAIL receptor expression on peripheral T lymphocytes in the molecular mechanism of RA development. METHODS: The expression of TRAIL/TRAIL receptors on T cells in 20 RA patients and 12 control individuals were analyzed using flow cytometry. The correlation of TRAIL and its receptor expression profile was compared with clinical RA parameters (RA activity scored as per DAS28) using Spearman Rho Analysis. RESULTS: While no change was detected in the ratio of CD4+ to CD8+ T cells between controls and RA patient groups, upregulation of TRAIL and its receptors (both death and decoy) was detected on both CD4+ and CD8+ T cells in RA patients compared to control individuals. Death Receptor-4 (DR4) and the decoy receptors DcR1 and DcR2 on CD8+ T cells, but not on CD4+ T cells, were positively correlated with patients' DAS scores. CONCLUSIONS: Our data suggest that TRAIL/TRAIL receptor expression profiles on T cells might be important in revelation of RA pathogenesis.


Assuntos
Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Linfócitos T CD8-Positivos/imunologia , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores Chamariz do Fator de Necrose Tumoral/biossíntese , Artrite Reumatoide/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Feminino , Proteínas Ligadas por GPI/biossíntese , Humanos , Masculino , Valor Preditivo dos Testes , Membro 10c de Receptores do Fator de Necrose Tumoral , Sensibilidade e Especificidade , Índice de Gravidade de Doença
17.
Methods Mol Biol ; 1879: 323-345, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29797007

RESUMO

Lentiviral vectors are becoming preferred vectors of choice for clinical gene therapy trials due to their safety, efficacy, and the long-term gene expression they provide. Although the efficacy of lentiviral vectors is mainly predetermined by the therapeutic genes they carry, they must be produced at high titers to exert therapeutic benefit for in vivo applications. Thus, there is need for practical, robust, and scalable viral vector production methods applicable to any laboratory setting. Here, we describe a practical lentiviral production technique in roller bottles yielding high-titer third-generation lentiviral vectors useful for in vivo gene transfer applications. CaPO4-mediated transient transfection protocol involving the use of a transfer vector and three different packaging plasmids is employed to generate lentivectors in roller bottles. Following clearance of cellular debris via low-speed centrifugation and filtration, virus is concentrated by high-speed ultracentrifugation over sucrose cushion.


Assuntos
Vetores Genéticos/genética , HIV/genética , Lentivirus/genética , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Células HEK293 , Humanos , Plasmídeos/genética , Transdução Genética/métodos , Transfecção/métodos , Ultracentrifugação/métodos
18.
Methods Mol Biol ; 1879: 347-365, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30006865

RESUMO

Lentiviral vectors (LVs) have been increasingly used in clinical gene therapy applications particularly due to their efficient gene transfer ability, lack of interference from preexisting viral immunity, and long-term gene expression they provide. Purity of LVs is essential in in vivo applications, for a high therapeutic benefit with minimum toxicity. Accordingly, laboratory scale production of LVs frequently involves transient cotransfection of 293T cells with packaging and transfer plasmids in the presence of CaPO4. After clearance of the cellular debris by low-speed centrifugation and filtration, lentivectors are usually concentrated by high-speed ultracentrifugation in sucrose cushion. Concentrated viral samples are then purified by anion exchange chromatography (AEX) after benzonase treatment to remove the residual cellular DNA. Here, we describe an improved practical method for LV purification using AEX, useful for experimental studies concerning gene and stem cell therapy.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Cromatografia por Troca Iônica/métodos , Terapia Genética , Vetores Genéticos/isolamento & purificação , HIV/genética , Lentivirus/genética , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Transdução Genética
19.
J Cell Biochem ; 104(3): 710-20, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18247339

RESUMO

Type 1 diabetes results from the T cell-mediated destruction of pancreatic beta cells. Islet transplantation has recently become a potential therapeutic approach for patients with type 1 diabetes. However, islet-graft failure appears to be a challenging issue to overcome. Thus, complementary gene therapy strategies are needed to improve the islet-graft survival following transplantation. Immune modulation through gene therapy represents a novel way of attacking cytotoxic T cells targeting pancreatic islets. Various death ligands of the TNF family such as FasL, TNF, and TNF-Related Apoptosis-Inducing Ligand (TRAIL) have been studied for this purpose. The over-expression of TNF or FasL in pancreatic islets exacerbates the onset of type 1 diabetes generating lymphocyte infiltrates responsible for the inflammation. Conversely, the lack of TRAIL expression results in higher degree of islet inflammation in the pancreas. In addition, blocking of TRAIL function using soluble TRAIL receptors facilitates the onset of diabetes. These results suggested that contrary to what was observed with TNF or FasL, adenovirus mediated TRAIL gene delivery into pancreatic islets is expected to be therapeutically beneficial in the setting of experimental models of type 1 diabetes. In conclusion; this study mainly reveals the fundamental principles of death ligand-mediated immune evasion in diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Terapia Genética/métodos , Sistema Imunitário/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Diabetes Mellitus Tipo 1/metabolismo , Proteína Ligante Fas/metabolismo , Técnicas de Transferência de Genes , Humanos , Ilhotas Pancreáticas/citologia , Ratos , Linfócitos T Citotóxicos/metabolismo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
20.
Urol Oncol ; 26(2): 158-65, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18312935

RESUMO

OBJECTIVE: Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has recently been investigated because of its ability to selectively kill cancer cells. Despite recent publications mainly focusing on TRAIL resistance in cancer cells, little is known about how TRAIL contributes to the carcinogenesis process. Because the expression patterns of TRAIL and its receptors in patients with prostate carcinoma have recently been reported, this study investigated the significance of TRAIL and TRAIL receptor expression in connection to serum prostate-specific antigen (PSA) and Gleason scoring. MATERIALS AND METHODS: A total of 98 patients were included in the study. Gleason scores, PSA, TRAIL, and TRAIL receptor expressions were used for the comparison purposes. The Spearman rho correlation test was administered to reveal the correlations among the variants. The Kruskal Wallis-Mann Whitney U or Friedman-Wilcoxon signed ranks test determined the statistical significance between the pairs. Multinomial and/or multiple binary logistic regression analyses were deployed to test whether TRAIL markers were independent variables to predict the prognosis of prostate cancer. Kaplan-Meier and log-rank tests were used to determine the survival rates. RESULTS: High-serum PSA levels were correlated with higher levels of TRAIL and TRAIL receptor expressions. Patients with high Gleason scores had higher levels of TRAIL-R4 decoy receptor expression but lower levels of TRAIL death ligand expression. CONCLUSIONS: TRAIL-R4 decoy receptor expression is strongly correlated with PSA recurrence, which is suggestive of poor prognosis. High levels of TRAIL-R4 expression but low levels of TRAIL death ligand expression are connected to decreased survival.


Assuntos
Antígeno Prostático Específico/sangue , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/mortalidade , Receptores Chamariz do Fator de Necrose Tumoral/biossíntese , Humanos , Masculino , Recidiva , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA