Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Genet Metab Rep ; 32: 100882, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35600090

RESUMEN

Phenylketonuria (PKU) is a genetic disorder affecting around 1 in 12,000 live births (1), caused by a mutation in the phenylalanine hydroxylase (PAH) gene in the liver which facilitates the catabolism of phenylalanine (Phe). Without a functional copy of PAH, levels of Phe in the blood and tissues rise, resulting in potentially life-threatening damage to the central nervous system. (2) Treatment options for PKU are limited, and center around adherence to a strict PKU diet that suffers from poor patient compliance. There are two approved drugs available, one of which must be used in conjunction with the PKU diet and another that has serious immunological side effects. Here we demonstrate that the LUNAR® delivery technology is capable of delivering mRNA for a replacement enzyme, the bacterial phenylalanine ammonia lyase (avPAL), into the hepatic tissue of a PKU mouse, and that the enzyme is capable of metabolizing Phe and reducing serum levels of Phe for more than five days post-transfection. We further demonstrate the ability of LUNAR to deliver a plant-derived PAL protein with a similar impact on the level of serum Phe. Taken together these results demonstrate both the capability of LUNAR for the targeted delivery of PAL mRNA into hepatic tissue in vivo, replacing the defective PAH protein and successfully reducing serum Phe levels, thereby addressing the underlying cause of PKU symptoms. Secondly, that plant-based PAL proteins are a viable alternative to bacterial avPAL to reduce the immunogenic response.

2.
Mol Metab ; 60: 101487, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35378329

RESUMEN

OBJECTIVE: Fibrotic organ responses have recently been identified as long-term complications in diabetes. Indeed, insulin resistance and aberrant hepatic lipid accumulation represent driving features of progressive non-alcoholic fatty liver disease (NAFLD), ranging from simple steatosis and non-alcoholic steatohepatitis (NASH) to fibrosis. Effective pharmacological regimens to stop progressive liver disease are still lacking to-date. METHODS: Based on our previous discovery of transforming growth factor beta-like stimulated clone (TSC)22D4 as a key driver of insulin resistance and glucose intolerance in obesity and type 2 diabetes, we generated a TSC22D4-hepatocyte specific knockout line (TSC22D4-HepaKO) and exposed mice to control or NASH diet models. Mechanistic insights were generated by metabolic phenotyping and single-nuclei RNA sequencing. RESULTS: Hepatic TSC22D4 expression was significantly correlated with markers of liver disease progression and fibrosis in both murine and human livers. Indeed, hepatic TSC22D4 levels were elevated in human NASH patients as well as in several murine NASH models. Specific genetic deletion of TSC22D4 in hepatocytes led to reduced liver lipid accumulation, improvements in steatosis and inflammation scores and decreased apoptosis in mice fed a lipogenic MCD diet. Single-nuclei RNA sequencing revealed a distinct TSC22D4-dependent gene signature identifying an upregulation of mitochondrial-related processes in hepatocytes upon loss of TSC22D4. An enrichment of genes involved in the TCA cycle, mitochondrial organization, and triglyceride metabolism underscored the hepatocyte-protective phenotype and overall decreased liver damage as seen in mouse models of hepatocyte-selective TSC22D4 loss-of-function. CONCLUSIONS: Together, our data uncover a new connection between targeted depletion of TSC22D4 and intrinsic metabolic processes in progressive liver disease. Hepatocyte-specific reduction of TSC22D4 improves hepatic steatosis and promotes hepatocyte survival via mitochondrial-related mechanisms thus paving the way for targeted therapies.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Diabetes Mellitus Tipo 2/metabolismo , Fibrosis , Hepatocitos/metabolismo , Humanos , Lípidos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factores de Transcripción/metabolismo
3.
Mol Ther Nucleic Acids ; 28: 87-98, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35356682

RESUMEN

Phenylketonuria (PKU) is an inborn error caused by deficiencies in phenylalanine (Phe) metabolism. Mutations in the phenylalanine hydroxylase (PAH) gene are the main cause of the disease whose signature hallmarks of toxically elevated levels of Phe accumulation in plasma and organs such as the brain, result in irreversible intellectual disability. Here, we present a unique approach to treating PKU deficiency by using an mRNA replacement therapy. A full-length mRNA encoding human PAH (hPAH) is encapsulated in our proprietary lipid nanoparticle LUNAR and delivered to a Pah enu2 mouse model that carries a missense mutation in the mouse PAH gene. Animals carrying this missense mutation develop hyperphenylalanemia and hypotyrosinemia in plasma, two clinical features commonly observed in the clinical presentation of PKU. We show that intravenous infusion of LUNAR-hPAH mRNA can generate high levels of hPAH protein in hepatocytes and restore the Phe metabolism in the Pah enu2 mouse model. Together, these data establish a proof of principle of a novel mRNA replacement therapy to treat PKU.

4.
Chem Phys Lipids ; 243: 105178, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35122738

RESUMEN

Lipid nanoparticles (LNPs) mediated mRNA delivery has gained prominence due to the success of mRNA vaccines against Covid-19, without which it would not have been possible. However, there is little clinical validation of this technology for other mRNA-based therapeutic approaches. Systemic administration of LNPs predominantly targets the liver, but delivery to other organs remains a challenge. Local approaches remain a viable option for some disease indications, such as Cystic Fibrosis, where aerosolized delivery to airway epithelium is the preferred route of administration. With this in mind, novel cationic lipids (L1-L4) have been designed, synthesized and co-formulated with a proprietary ionizable lipid. These LNPs were further nebulized, along with baseline control DOTAP-based LNP (DOTAP+), and tested in vitro for mRNA integrity and encapsulation efficiency, as well as transfection efficiency and cytotoxicity in cell cultures. Improved biodegradability and potentially superior elimination profiles of L1-L4, in part due to physicochemical characteristics of putative metabolites, are thought to be advantageous for prospective therapeutic lung delivery applications using these lipids.


Asunto(s)
Liposomas/química , Pulmón , Nanopartículas/química , ARN Mensajero/administración & dosificación , Humanos
5.
Mol Ther Nucleic Acids ; 24: 1-10, 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-33738134

RESUMEN

Polyglutamine (polyQ) diseases are inherited neurodegenerative disorders caused by expansion of cytosine-adenine-guanine (CAG)-trinucleotide repeats in causative genes. These diseases include spinal and bulbar muscular atrophy (SBMA), Huntington's disease, dentatorubral-pallidoluysian atrophy, and spinocerebellar ataxias. Targeting expanded CAG repeats is a common therapeutic approach to polyQ diseases, but concomitant silencing of genes with normal CAG repeats may lead to toxicity. Previous studies have shown that CAG repeat-targeting small interfering RNA duplexes (CAG-siRNAs) have the potential to selectively suppress mutant proteins in in vitro cell models of polyQ diseases. However, in vivo application of these siRNAs has not yet been investigated. In this study, we demonstrate that an unlocked nucleic acid (UNA)-modified CAG-siRNA shows high selectivity for polyQ-expanded androgen receptor (AR) inhibition in in vitro cell models and that lipid nanoparticle (LNP)-mediated delivery of the CAG-siRNA selectively suppresses mutant AR in the central nervous system of an SBMA mouse model. In addition, a subcutaneous injection of the LNP-delivered CAG-siRNA efficiently suppresses mutant AR in the skeletal muscle of the SBMA mouse model. These results support the therapeutic potential of LNP-delivered UNA-modified CAG-siRNAs for selective suppression of mutant proteins in SBMA and other polyQ diseases.

6.
Sci Adv ; 6(45)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33148638

RESUMEN

Zika virus (ZIKV) is associated with congenital malformations in infants born to infected mothers, and with Guillain-Barré syndrome in infected adults. Development of ZIKV vaccines has focused predominantly on the induction of neutralizing antibodies, although a suboptimal antibody response may theoretically enhance disease severity through antibody-dependent enhancement (ADE). Here, we report induction of a protective anti-ZIKV CD8+ T cell response in the HLA-B*0702 Ifnar1-/- transgenic mice using an alphavirus-based replicon RNA vaccine expressing ZIKV nonstructural protein NS3, a potent T cell antigen. The NS3 vaccine did not induce a neutralizing antibody response but elicited polyfunctional CD8+ T cells that were necessary and sufficient for preventing death in lethally infected adult mice and fetal growth restriction in infected pregnant mice. These data identify CD8+ T cells as the major mediators of ZIKV NS3 vaccine-induced protection and suggest a new strategy to develop safe and effective anti-flavivirus vaccines.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Animales , Anticuerpos Neutralizantes , Linfocitos T CD8-positivos , Humanos , Ratones , Vacunas Sintéticas , Vacunas de ARNm
7.
J Med Chem ; 63(21): 12992-13012, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33119286

RESUMEN

Ionizable cationic lipids are critical components involved in nanoparticle formulations, which are utilized in delivery platforms for RNA therapeutics. While general criteria regarding lipophilicity and measured pKa in formulation are understood to have impacts on utility in vivo, greater granularity with respect to the impacts of the structure on calculated and measured physicochemical parameters and the subsequent performance of those ionizable cationic lipids in in vivo studies would be beneficial. Herein, we describe structural alterations made within a lipid class exemplified by 4, which allow us to tune calculated and measured physicochemical parameters for improved performance, resulting in substantial improvements versus the state of the art at the outset of these studies, resulting in good in vivo activity within a range of measured basicity (pKa = 6.0-6.6) and lipophilicity (cLogD = 10-14).


Asunto(s)
Lípidos/química , ARN Interferente Pequeño/metabolismo , Transfección/métodos , Animales , Cationes/química , Factor VII/antagonistas & inhibidores , Factor VII/genética , Factor VII/metabolismo , Femenino , Humanos , Cinética , Lípidos/síntesis química , Ratones , Nanopartículas/química , Tamaño de la Partícula , Interferencia de ARN , Estabilidad del ARN , ARN Interferente Pequeño/sangre , Relación Estructura-Actividad
8.
Front Microbiol ; 11: 832, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508764

RESUMEN

We explored an emerging technology to produce anti-Hantaan virus (HTNV) and anti-Puumala virus (PUUV) neutralizing antibodies for use as pre- or post-exposure prophylactics. The technology involves hyperimmunization of transchomosomic bovines (TcB) engineered to express human polyclonal IgG antibodies with HTNV and PUUV DNA vaccines encoding GnGc glycoproteins. For the anti-HTNV product, TcB was hyperimmunized with HTNV DNA plus adjuvant or HTNV DNA formulated using lipid nanoparticles (LNP). The LNP-formulated vaccine yielded fivefold higher neutralizing antibody titers using 10-fold less DNA. Human IgG purified from the LNP-formulated animal (SAB-159), had anti-HTNV neutralizing antibody titers >100,000. SAB-159 was capable of neutralizing pseudovirions with monoclonal antibody escape mutations in Gn and Gc demonstrating neutralization escape resistance. SAB-159 protected hamsters from HTNV infection when administered pre- or post-exposure, and limited HTNV infection in a marmoset model. An LNP-formulated PUUV DNA vaccine generated purified anti-PUUV IgG, SAB-159P, with a neutralizing antibody titer >600,000. As little as 0.33 mg/kg of SAB-159P protected hamsters against PUUV infection for pre-exposure and 10 mg/kg SAB-159P protected PUUV-infected hamsters post-exposure. These data demonstrate that DNA vaccines combined with the TcB-based manufacturing platform can be used to rapidly produce potent, human, polyclonal, escape-resistant anti-HTNV, and anti-PUUV neutralizing antibodies that are protective in animal models.

9.
Neuro Oncol ; 19(3): 372-382, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27765835

RESUMEN

Background: Previously we showed therapeutic efficacy of unprotected miR-124 in preclinical murine models of glioblastoma, including in heterogeneous genetically engineered murine models by exploiting the immune system and thereby negating the need for direct tumor delivery. Although these data were promising, to implement clinical trials, we required a scalable formulation that afforded protection against circulatory RNases. Methods: We devised lipid nanoparticles that encapsulate and protect the miRs from degradation and provide enhanced delivery into the immune cell compartment and tested in vivo antitumor effects. Results: Treatment with nanoparticle-encapsulated miR-124, LUNAR-301, demonstrated a median survival exceeding 70 days, with an associated reversal of tumor-mediated immunosuppression and induction of immune memory. In both canine and murine models, the safety profile of LUNAR-301 was favorable. Conclusions: For the first time, we show that nanoparticles can direct a therapeutic response by targeting intracellular immune pathways. Although shown in the context of gliomas, this therapeutic approach would be applicable to other malignancies.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Tolerancia Inmunológica/genética , Lípidos/química , MicroARNs/genética , Nanopartículas/administración & dosificación , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Perros , Glioma/genética , Glioma/inmunología , Humanos , Memoria Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/administración & dosificación , Nanopartículas/química , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Pharm Res ; 30(2): 342-51, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22983644

RESUMEN

PURPOSE: To determine the influence of physicochemical properties of lipid nanoparticles (LNPs) carrying siRNA on their gene silencing in vivo. Mechanistic understanding of how the architecture of the nanoparticle can alter gene expression has also been studied. METHODS: The effect of 3-N-[(ω-methoxypoly(ethylene glycol)2000)carbamoyl]-1,2-dimyristyloxy-propylamine (PEG-C-DMA) on hepatic distribution and FVII gene silencing was determined. FVII mRNA in hepatocytes and liver tissues was determined by Q-PCR. Hepatic distribution was quantified by FACS analysis using Cy5 labeled siRNA. RESULTS: Gene silencing was highly dependent on the amount of PEG-C-DMA present. FVII gene silencing inversely correlated to the amount of PEG-C-DMA in LNPs. High FVII gene silencing was obtained in vitro and in vivo when the molar ratio of PEG-C-DMA to lipid was 0.5 mol%. Surprisingly, PEGylation didn't alter the hepatic distribution of the LNPs at 5 h post administration. Instead the amount of PEG present in the LNPs has an effect on red blood cell disruption at low pH. CONCLUSION: Low but sufficient PEG-C-DMA amount in LNPs plays an important role for efficient FVII gene silencing in vivo. PEGylation did not alter the hepatic distribution of LNPs, but altered gene silencing efficacy by potentially reducing endosomal disruption.


Asunto(s)
Factor VII/genética , Hepatocitos/metabolismo , Nanopartículas/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Animales , Células Cultivadas , Lípidos/química , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Propilaminas/química , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacocinética , Ratas , Ratas Sprague-Dawley
11.
J Control Release ; 161(1): 124-31, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22521595

RESUMEN

Polymeric prodrugs are one of the most promising chemotherapeutic agent delivery approaches, displaying unique drug release profiles, serum stability, formulation flexibility, and reduced drug resistance. One of the most important aspects of a polymeric prodrug, albeit a less-extensively studied one, is the polymer's molecular weight, which affects particle formation, drug release and PK/PD profiles, drug stability, and cell uptake; these factors in turn affect the prodrug's maximum tolerated dose and anticancer efficacy. Poly(L-γ-glutamylglutamine) (PGG) is a linear polymer designed to improve the therapeutic index of attached drugs. In this study we selected poly(L-γ-glutamylglutamine)-paclitaxel (PGG­PTX), as a model system for the methodical investigation into the effects of the poly(L-γ-glutamylglutamine) backbone molecular weight on its pharmacological performance. The polymeric prodrug was characterized by NMR, DLS and GPC-MALS, and its anticancer activity in vitro and in vivo was assessed. Herein we present data which provide valuable insight into improving anticancer polymer-based prodrug design and development.


Asunto(s)
Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Paclitaxel/análogos & derivados , Proteínas/química , Proteínas/uso terapéutico , Animales , Antineoplásicos Fitogénicos/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Peso Molecular , Neoplasias/tratamiento farmacológico , Paclitaxel/química , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Profármacos/química , Profármacos/farmacocinética , Profármacos/uso terapéutico , Proteínas/farmacocinética
12.
J Control Release ; 140(3): 277-83, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19699770

RESUMEN

BACKGROUND: Targeting of a specific subset of cells is mandatory for the successful application of siRNA mediated silencing in anticancer therapy. A recent theory suggests that colon cancer is sustained by a small subpopulation of cells, termed cancer stem cells (CSCs). These cells are characterized by their innate drug resistance properties, which is one of the key factors of chemotherapy failure. The goal of this study was to assess whether a novel siRNA delivery carrier, with an appropriate siRNA, targeted to CD133+ cells has the potential to improve the efficacy of conventional chemotherapy. METHODS: In this study, a novel synthetic siRNA carrier platform was designed and synthesized. This carrier was composed of a cationic oligomer (PEI(1200)), a hydrophilic polymer (polyethylene glycol) and a biodegradable lipid-based crosslinking moiety. Libraries of polymers were synthesized by varying their lipid composition. Their transfection efficacy was evaluated in vitro using CHOK1 cells. The polymer was characterized using molecular weight, particle encapsulation assay, particle size and surface charge analysis. RESULTS: It was demonstrated that the lipid composition in the polymer plays a critical role in transfection. Optimizing the physicochemical properties of the polymers is crucial in achieving favorable knockdown. Lipid nano complex with composition PEI-Lipid(1:16) was the optimum ratio for gene silencing. Additionally, silencing of multidrug resistance gene (MDR1) and treatment with paclitaxel play a synergistic role in increasing the efficacy as compared to the drug alone. CONCLUSIONS: In the present study a novel siRNA delivery carrier system with an MDR1-targeting siRNA (siMDR1) effectively reduced the expression of MDR1 in human colon CSCs (CD133+ enriched cell population), resulting in significantly increasing the chemosensitivity to paclitaxel.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Paclitaxel/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/uso terapéutico , Antígeno AC133 , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adyuvantes Farmacéuticos/farmacología , Antígenos CD/metabolismo , Antineoplásicos Fitogénicos/farmacología , Cationes , Línea Celular Tumoral , Neoplasias del Colon/patología , Reactivos de Enlaces Cruzados , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Glicoproteínas/metabolismo , Humanos , Lípidos/química , Luciferasas/genética , Peso Molecular , Paclitaxel/farmacología , Tamaño de la Partícula , Péptidos/metabolismo , Polietilenglicoles/química , Polímeros , Solubilidad , Transfección , Agua
13.
Biomaterials ; 27(7): 1140-51, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16098577

RESUMEN

Novel interpenetrating network (IPN) hydrogels, composed of pH-sensitive, aromatic azo group containing network as one component (Network A), and a hydrolyzable network as the other (Network B), were prepared by a sequential process. The first network was formed by crosslinking of a reactive polymer precursor (copolymer of N,N-dimethylacrylamide, acrylic acid, N-tert.butylacrylamide, and N-methacryloylglycylglycine p-nitrophenyl ester) with an aromatic azo group containing diamine ((N,N'-epsilon-aminocaproyl)-4,4'-diaminoazobenzene). The second network was formed by radical crosslinking copolymerization of N-(2-hydroxypropyl)methacrylamide with N,O-dimethacryloylhydroxylamine. The composition of the hydrogels was manipulated to determine the influence of hydrogel composition on the equilibrium degree of swelling, modulus of elasticity in compression, and on the rate of degradation of Network B. Modeling of network structure was accomplished using the statistical branching theory. The major advantage of IPN hydrogels, when compared to traditional pH-sensitive networks, is the linear swelling profile following abrupt change of pH from 2 to 7.4. This indicates the suitability of IPN as carriers for oral drug delivery.


Asunto(s)
Compuestos Azo/química , Materiales Biocompatibles/química , Portadores de Fármacos/química , Hidrocarburos Aromáticos/química , Hidrogeles/química , Absorción , Elasticidad , Concentración de Iones de Hidrógeno , Hidrólisis , Ensayo de Materiales , Conformación Molecular , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...