Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 389(7): 620-631, 2023 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-37585628

RESUMEN

BACKGROUND: Patients with the Crigler-Najjar syndrome lack the enzyme uridine diphosphoglucuronate glucuronosyltransferase 1A1 (UGT1A1), the absence of which leads to severe unconjugated hyperbilirubinemia that can cause irreversible neurologic injury and death. Prolonged, daily phototherapy partially controls the jaundice, but the only definitive cure is liver transplantation. METHODS: We report the results of the dose-escalation portion of a phase 1-2 study evaluating the safety and efficacy of a single intravenous infusion of an adeno-associated virus serotype 8 vector encoding UGT1A1 in patients with the Crigler-Najjar syndrome that was being treated with phototherapy. Five patients received a single infusion of the gene construct (GNT0003): two received 2×1012 vector genomes (vg) per kilogram of body weight, and three received 5×1012 vg per kilogram. The primary end points were measures of safety and efficacy; efficacy was defined as a serum bilirubin level of 300 µmol per liter or lower measured at 17 weeks, 1 week after discontinuation of phototherapy. RESULTS: No serious adverse events were reported. The most common adverse events were headache and alterations in liver-enzyme levels. Alanine aminotransferase increased to levels above the upper limit of the normal range in four patients, a finding potentially related to an immune response against the infused vector; these patients were treated with a course of glucocorticoids. By week 16, serum bilirubin levels in patients who received the lower dose of GNT0003 exceeded 300 µmol per liter. The patients who received the higher dose had bilirubin levels below 300 µmol per liter in the absence of phototherapy at the end of follow-up (mean [±SD] baseline bilirubin level, 351±56 µmol per liter; mean level at the final follow-up visit [week 78 in two patients and week 80 in the other], 149±33 µmol per liter). CONCLUSIONS: No serious adverse events were reported in patients treated with the gene-therapy vector GNT0003 in this small study. Patients who received the higher dose had a decrease in bilirubin levels and were not receiving phototherapy at least 78 weeks after vector administration. (Funded by Genethon and others; ClinicalTrials.gov number, NCT03466463.).


Asunto(s)
Síndrome de Crigler-Najjar , Terapia Genética , Glucuronosiltransferasa , Humanos , Administración Intravenosa , Bilirrubina/sangre , Síndrome de Crigler-Najjar/sangre , Síndrome de Crigler-Najjar/complicaciones , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/terapia , Dependovirus , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glucuronosiltransferasa/administración & dosificación , Glucuronosiltransferasa/genética , Hiperbilirrubinemia/sangre , Hiperbilirrubinemia/etiología , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/terapia , Trasplante de Hígado , Fototerapia
2.
J Inherit Metab Dis ; 44(6): 1382-1392, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34528713

RESUMEN

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of sulfur metabolism characterized by elevated blood levels of total homocysteine (tHcy). Patients diagnosed with CBS deficiency are currently treated by a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine, but the effectiveness of this therapy is limited due to poor compliance. A mouse model for CBS deficiency (Tg-I278T Cbs-/- ) was used to evaluate a potential gene therapy approach to treat CBS deficiency utilizing an AAVrh.10-based vector containing the human CBS cDNA downstream of the constitutive, strong CAG promoter (AAVrh.10hCBS). Mice were administered a single dose of virus and followed for up to 1 year. The data demonstrated a dose-dependent increase in liver CBS activity and a dose-dependent decrease in serum tHcy. Liver CBS enzyme activity at 1 year was similar to Cbs+/- control mice. Mice given the highest dose (5.6 × 1011 genomes/mouse) had mean serum tHcy decrease of 97% 1 week after injection and an 81% reduction 1 year after injection. Treated mice had either full- or substantial correction of alopecia, bone loss, and fat mass phenotypes associated with Cbs deficiency in mice. Our findings show that AAVrh.10-based gene therapy is highly effective in treating CBS deficiency in mice and supports additional pre-clinical testing for eventual use human trials.


Asunto(s)
Cistationina betasintasa/genética , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Homocistinuria/genética , Homocistinuria/terapia , Animales , Cistationina betasintasa/sangre , Cistationina betasintasa/deficiencia , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Homocistinuria/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Fenotipo
3.
Mol Genet Metab ; 134(1-2): 139-146, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34454844

RESUMEN

Maple syrup urine disease (MSUD) is a rare, inherited metabolic disorder characterized by a dysfunctional mitochondrial enzyme complex, branched-chain alpha-keto acid dehydrogenase (BCKDH), which catabolizes branched-chain amino acids (BCAAs). Without functional BCKDH, BCAAs and their neurotoxic alpha-keto intermediates can accumulate in the blood and tissues. MSUD is currently incurable and treatment is limited to dietary restriction or liver transplantation, meaning there is a great need to develop new treatments for MSUD. We evaluated potential gene therapy applications for MSUD in the intermediate MSUD (iMSUD) mouse model, which harbors a mutation in the dihydrolipoamide branched-chain transacylase E2 (DBT) subunit of BCKDH. Systemic delivery of an adeno-associated virus (AAV) vector expressing DBT under control of the liver-specific TBG promoter to the liver did not sufficiently ameliorate all aspects of the disease phenotype. These findings necessitated an alternative therapeutic strategy. Muscle makes a larger contribution to BCAA metabolism than liver in humans, but a muscle-specific approach involving a muscle-specific promoter for DBT expression delivered via intramuscular (IM) administration only partially rescued the MSUD phenotype in mice. Combining the muscle-tropic AAV9 capsid with the ubiquitous CB7 promoter via IM or IV injection, however, substantially increased survival across all assessed doses. Additionally, near-normal serum BCAA levels were achieved and maintained in the mid- and high-dose cohorts throughout the study; this approach also protected these mice from a lethal high-protein diet challenge. Therefore, administration of a gene therapy vector that expresses in both muscle and liver may represent a viable approach to treating patients with MSUD.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Enfermedad de la Orina de Jarabe de Arce/genética , Enfermedad de la Orina de Jarabe de Arce/terapia , Fenotipo , Administración Intravenosa , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Masculino , Ratones , Mutación
4.
Viruses ; 13(6)2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208264

RESUMEN

Oncolytic virotherapy (OV) is an emerging class of immunotherapeutic drugs. Their mechanism of action is two-fold: direct cell lysis and unmasking of the cancer through immunogenic cell death, which allows the immune system to recognize and eradicate tumours. Breast cancer is the most common cancer in women and is challenging to treat with immunotherapy modalities because it is classically an immunogenically "cold" tumour type. This provides an attractive niche for OV, given viruses have been shown to turn "cold" tumours "hot," thereby opening a plethora of treatment opportunities. There has been a number of pre-clinical attempts to explore the use of OV in breast cancer; however, these have not led to any meaningful clinical trials. This review considers both the potential and the barriers to OV in breast cancer, namely, the limitations of monotherapy and the scope for combination therapy, improving viral delivery and challenges specific to the breast cancer population (e.g., tumour subtype, menopausal status, age).


Asunto(s)
Neoplasias de la Mama/terapia , Terapia Genética , Viroterapia Oncolítica , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/etiología , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Femenino , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Resultado del Tratamiento
5.
Sci Rep ; 11(1): 10400, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34002008

RESUMEN

The lateral hypothalamus (LH) is critically involved in the regulation of homeostatic energy balance. Some neurons in the LH express receptors for leptin (LepRb), a hormone known to increase energy expenditure and decrease energy intake. However, the neuroanatomical inputs to LepRb-expressing LH neurons remain unknown. We used rabies virus tracing technology to map these inputs, but encountered non-specific tracing. To optimize this technology for a minor cell population (LepRb is not ubiquitously expressed in LH), we used LepRb-Cre mice and assessed how different titers of the avian tumor virus receptor A (TVA) helper virus affected rabies tracing efficiency and specificity. We found that rabies expression is dependent on TVA receptor expression, and that leakiness of TVA receptors is dependent on the titer of TVA virus used. We concluded that a titer of 1.0-3.0 × 107 genomic copies per µl of the TVA virus is optimal for rabies tracing. Next, we successfully applied modified rabies virus tracing technology to map inputs to LepRb-expressing LH neurons. We discovered that other neurons in the LH itself, the periventricular hypothalamic nucleus (Pe), the posterior hypothalamic nucleus (PH), the bed nucleus of the stria terminalis (BNST), and the paraventricular hypothalamic nucleus (PVN) are the most prominent input areas to LepRb-expressing LH neurons.


Asunto(s)
Conectoma/métodos , Hipotálamo/diagnóstico por imagen , Imagen Molecular/métodos , Neuronas/metabolismo , Receptores de Leptina/análisis , Animales , Proteínas Aviares/genética , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Virus Helper/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Virus de la Rabia/genética , Receptores de Leptina/metabolismo , Receptores Virales/genética , Núcleos Septales/citología , Núcleos Septales/diagnóstico por imagen , Núcleos Septales/metabolismo , Técnicas Estereotáxicas
6.
Front Immunol ; 12: 658038, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868303

RESUMEN

Recombinant adeno-associated virus (rAAV) has attracted a significant research focus for delivering genetic therapies to target cells. This non-enveloped virus has been trialed in many clinical-stage therapeutic strategies but important obstacle in clinical translation is the activation of both innate and adaptive immune response to the protein capsid, vector genome and transgene product. In addition, the normal population has pre-existing neutralizing antibodies against wild-type AAV, and cross-reactivity is observed between different rAAV serotypes. While extent of response can be influenced by dosing, administration route and target organ(s), these pose concerns over reduction or complete loss of efficacy, options for re-administration, and other unwanted immunological sequalae such as local tissue damage. To reduce said immunological risks, patients are excluded if they harbor anti-AAV antibodies or have received gene therapy previously. Studies have incorporated immunomodulating or suppressive regimens to block cellular and humoral immune responses such as systemic corticosteroids pre- and post-administration of Luxturna® and Zolgensma®, the two rAAV products with licensed regulatory approval in Europe and the United States. In this review, we will introduce the current pharmacological strategies to immunosuppress or immunomodulate the host immune response to rAAV gene therapy.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Inmunomodulación , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Inmunidad Celular , Inmunidad Humoral , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Inmunomodulación/efectos de los fármacos , Transgenes/genética
7.
Mol Ther ; 29(8): 2499-2513, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-33839322

RESUMEN

Recurrent episodes of decompensated heart failure (HF) represent an emerging cause of hospitalizations in developed countries with an urgent need for effective therapies. Recently, the pregnancy-related hormone relaxin (RLN) was found to mediate cardio-protective effects and act as a positive inotrope in the cardiovascular system. RLN binds to the RLN family peptide receptor 1 (RXFP1), which is predominantly expressed in atrial cardiomyocytes. We therefore hypothesized that ventricular RXFP1 expression might exert potential therapeutic effects in an in vivo model of cardiac dysfunction. Thus, mice were exposed to pressure overload by transverse aortic constriction and treated with AAV9 to ectopically express RXFP1. To activate RXFP1 signaling, RLN was supplemented subcutaneously. Ventricular RXFP1 expression was well tolerated. Additional RLN administration not only abrogated HF progression but restored left ventricular systolic function. In accordance, upregulation of fetal genes and pathological remodeling markers were significantly reduced. In vitro, RLN stimulation of RXFP1-expressing cardiomyocytes induced downstream signaling, resulting in protein kinase A (PKA)-specific phosphorylation of phospholamban (PLB), which was distinguishable from ß-adrenergic activation. PLB phosphorylation corresponded to increased calcium amplitude and contractility. In conclusion, our results demonstrate that ligand-activated cardiac RXFP1 gene therapy represents a therapeutic approach to attenuate HF with the potential to adjust therapy by exogenous RLN supplementation.


Asunto(s)
Terapia Genética/métodos , Insuficiencia Cardíaca/terapia , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Relaxina/administración & dosificación , Animales , Proteínas de Unión al Calcio/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Inyecciones Subcutáneas , Ligandos , Masculino , Ratones , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Resultado del Tratamiento , Función Ventricular
8.
Theranostics ; 11(2): 649-664, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391497

RESUMEN

The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a worldwide epidemic of the lethal respiratory coronavirus disease (COVID-19), necessitating urgent development of specific and effective therapeutic tools. Among several therapeutic targets of coronaviruses, the spike protein is of great significance due to its key role in host invasion. Here, we report a potential anti-SARS-CoV-2 strategy based on the CRISPR-Cas13a system. Methods: A comprehensive set of bioinformatics methods, including sequence alignment, structural comparison, and molecular docking, was utilized to identify a SARS-CoV-2-spike(S)-specific segment. A tiling crRNA library targeting this specific RNA segment was designed, and optimal crRNA candidates were selected using in-silico methods. The efficiencies of the crRNA candidates were tested in human HepG2 and AT2 cells. Results: The most effective crRNA sequence inducing a robust cleavage effect on S and a potent collateral cleavage effect were identified. Conclusions: This study provides a rapid design pipeline for a CRISPR-Cas13a-based antiviral tool against SARS-CoV-2. Moreover, it offers a novel approach for anti-virus study even if the precise structures of viral proteins are indeterminate.


Asunto(s)
Antivirales/administración & dosificación , Tratamiento Farmacológico de COVID-19 , ARN Guía de Kinetoplastida/genética , Glicoproteína de la Espiga del Coronavirus/genética , COVID-19/virología , Sistemas CRISPR-Cas/genética , Biología Computacional , Evaluación Preclínica de Medicamentos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células Hep G2 , Humanos , Simulación del Acoplamiento Molecular , SARS-CoV-2/genética , Homología de Secuencia de Aminoácido
9.
Nat Commun ; 12(1): 697, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514733

RESUMEN

Mutations in voltage-gated potassium channel KCNE1 cause Jervell and Lange-Nielsen syndrome type 2 (JLNS2), resulting in congenital deafness and vestibular dysfunction. We conducted gene therapy by injecting viral vectors using the canalostomy approach in Kcne1-/- mice to treat both the hearing and vestibular symptoms. Results showed early treatment prevented collapse of the Reissner's membrane and vestibular wall, retained the normal size of the semicircular canals, and prevented the degeneration of inner ear cells. In a dose-dependent manner, the treatment preserved auditory (16 out of 20 mice) and vestibular (20/20) functions in mice treated with the high-dosage for at least five months. In the low-dosage group, a subgroup of mice (13/20) showed improvements only in the vestibular functions. Results supported that highly efficient transduction is one of the key factors for achieving the efficacy and maintaining the long-term therapeutic effect. Secondary outcomes of treatment included improved birth and litter survival rates. Our results demonstrated that gene therapy via the canalostomy approach, which has been considered to be one of the more feasible delivery methods for human inner ear gene therapy, preserved auditory and vestibular functions in a dose-dependent manner in a mouse model of JLNS2.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Síndrome de Jervell-Lange Nielsen/terapia , Canales de Potasio con Entrada de Voltaje/genética , Canales Semicirculares/cirugía , Animales , Animales Recién Nacidos , Dependovirus , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Audición/genética , Humanos , Inyecciones/métodos , Síndrome de Jervell-Lange Nielsen/genética , Masculino , Ratones , Ratones Noqueados , Parvovirinae/genética , Propiocepción/genética
10.
Adv Drug Deliv Rev ; 168: 99-117, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32931860

RESUMEN

Genome-editing tools such as Cre recombinase (Cre), zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and most recently the clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein system have revolutionized biomedical research, agriculture, microbial engineering, and therapeutic development. Direct delivery of genome editing enzymes, as opposed to their corresponding DNA and mRNA precursors, is advantageous since they do not require transcription and/or translation. In addition, prolonged overexpression is a problem when delivering viral vector or plasmid DNA which is bypassed when delivering whole proteins. This lowers the risk of insertional mutagenesis and makes for relatively easier manufacturing. However, a major limitation of utilizing genome editing proteins in vivo is their low delivery efficiency, and currently the most successful strategy involves using potentially immunogenic viral vectors. This lack of safe and effective non-viral delivery systems is still a big hurdle for the clinical translation of such enzymes. This review discusses the challenges of non-viral delivery strategies of widely used genome editing enzymes, including Cre recombinase, ZFNs and TALENs, CRISPR/Cas9, and Cas12a (Cpf1) in their protein format and highlights recent innovations of non-viral delivery strategies which have the potential to overcome current delivery limitations and advance the clinical translation of genome editing.


Asunto(s)
Edición Génica/métodos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Proteínas Bacterianas/administración & dosificación , Proteínas Asociadas a CRISPR/administración & dosificación , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Dendrímeros/química , Endodesoxirribonucleasas/administración & dosificación , Oro/química , Integrasas/administración & dosificación , Lípidos/química , Nanopartículas/química , Fósforo/química , Polietileneimina/química , Nucleasas de los Efectores Tipo Activadores de la Transcripción/administración & dosificación , Nucleasas con Dedos de Zinc/administración & dosificación
11.
Mol Ther ; 29(2): 597-610, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33309883

RESUMEN

Evaluation of immune responses to adeno-associated virus (AAV)-mediated gene therapies prior to and following dose administration plays a key role in determining therapeutic safety and efficacy. This report describes up to 3 years of immunogenicity data following administration of valoctocogene roxaparvovec (BMN 270), an AAV5-mediated gene therapy encoding human B domain-deleted FVIII (hFVIII-SQ) in a phase 1/2 clinical study of adult males with severe hemophilia A. Patients with pre-existing humoral immunity to AAV5 or with a history of FVIII inhibitors were excluded from the trial. Blood plasma and peripheral blood mononuclear cell (PBMC) samples were collected at regular intervals following dose administration for assessment of humoral and cellular immune responses to both the AAV5 vector and transgene-expressed hFVIII-SQ. The predominant immune response elicited by BMN 270 administration was largely limited to the development of antibodies against the AAV5 capsid that were cross-reactive with other common AAV serotypes. No FVIII inhibitor responses were observed within 3 years following dose administration. In a context of prophylactic or on-demand corticosteroid immunosuppression given after vector infusion, AAV5 and hFVIII-SQ peptide-specific cellular immune responses were intermittently detected by an interferon (IFN)-γ and tumor necrosis factor (TNF)-α FluoroSpot assay, but they were not clearly associated with detrimental safety events or changes in efficacy measures.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Hemofilia A/genética , Hemofilia A/terapia , Adulto , Reacciones Cruzadas/inmunología , Dependovirus/inmunología , Factor VIII/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Humoral , Masculino , Transgenes , Resultado del Tratamiento
12.
Int J Mol Sci ; 21(20)2020 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-33050329

RESUMEN

Malignant brain tumors remain incurable diseases. Although much effort has been devoted to improving patient outcome, multiple factors such as the high tumor heterogeneity, the strong tumor-induced immunosuppressive microenvironment, and the low mutational burden make the treatment of these tumors especially challenging. Thus, novel therapeutic strategies are urgent. Oncolytic viruses (OVs) are biotherapeutics that have been selected or engineered to infect and selectively kill cancer cells. Increasingly, preclinical and clinical studies demonstrate the ability of OVs to recruit T cells and induce durable immune responses against both virus and tumor, transforming a "cold" tumor microenvironment into a "hot" environment. Besides promising clinical results as a monotherapy, OVs can be powerfully combined with other cancer therapies, helping to overcome critical barriers through the creation of synergistic effects in the fight against brain cancer. Although many questions remain to be answered to fully exploit the therapeutic potential of OVs, oncolytic virotherapy will clearly be part of future treatments for patients with malignant brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Vectores Genéticos/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Biomarcadores de Tumor , Ensayos Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Viroterapia Oncolítica/métodos , Transducción Genética , Resultado del Tratamiento
13.
J Invest Dermatol ; 140(12): 2371-2379, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32335129

RESUMEN

Targeting neuroendocrine receptors can be considered as another interesting approach to treating fibrotic disorders. Previously, we could demonstrate that tropisetron, a classical serotonin receptor blocker, can modulate collagen synthesis and acts in vitro through the α7 nicotinic acetylcholine receptor (α7nAchR). Here, we used a pharmacologic approach with specific α7nAchR agonists to validate this hypothesis. PHA-543613, an α7nAchR-specific agonist, not only prevented but also reversed established skin fibrosis of mice injected with bleomycin. Interestingly, agonistic stimulation of α7nAchR also attenuated experimental skin fibrosis in the non-inflammation driven adenovirus coding for TGFß receptor Iact mouse model, indicating fibroblast-mediated and not only anti-inflammatory effects of such agents. The fibroblast-mediated effects were confirmed in vitro using human dermal fibroblasts, in which the α7nAchR-specific agonists strongly reduced the impact of TGFß1-mediated expression on collagen and myofibroblast marker expression. These actions were linked to modulation of the redox-sensitive transcription factor JunB and impairment of the mitochondrial respiratory system. Our results indicate that pharmacologic stimulation of the α7nAchR could be a promising target for treatment of patients with skin fibrotic diseases. Moreover, our results suggest a mechanistic axis of collagen synthesis regulation through the mitochondrial respiratory system.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Quinuclidinas/farmacología , Esclerodermia Sistémica/tratamiento farmacológico , Piel/patología , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Adenoviridae/genética , Animales , Bleomicina/toxicidad , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Cultivo Primario de Células , Quinuclidinas/uso terapéutico , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Piel/citología , Piel/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
14.
Hum Gene Ther ; 31(1-2): 70-79, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31650869

RESUMEN

Cocaine addiction continues to impose major burdens on affected individuals and broader society but is highly resistant to medical treatment or psychotherapy. This study was undertaken with the goal of Food and Drug Administration (FDA) permission for a first-in-human clinical trial of a gene therapy for treatment-seeking cocaine users to become and remain abstinent. The approach was based on intravenous administration of AAV8-hCocH, an adeno-associated viral vector encoding a modified plasma enzyme that metabolizes cocaine into harmless by-products. To assess systemic safety, we conducted "Good Laboratory Practice" (GLP) studies in cocaine-experienced and cocaine-naive mice at doses of 5E12 and 5E13 vector genomes/kg. Results showed total lack of viral vector-related adverse effects in all tests performed. Instead, mice given one injection of AAV8-hCocH and regular daily injections of cocaine had far less tissue pathology than cocaine-injected mice with no vector treatment. Biodistribution analysis showed the vector located almost exclusively in the liver. These results indicate that a liver-directed AAV8-hCocH gene transfer at reasonable dosage is safe, well tolerated, and effective. Thus, gene transfer therapy emerges as a radically new approach to treat compulsive cocaine abuse. In fact, based on these positive findings, the FDA recently accepted our latest request for investigational new drug application (IND 18579).


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Proteínas Recombinantes/genética , Animales , Biomarcadores , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/terapia , Dependovirus/clasificación , Susceptibilidad a Enfermedades , Evaluación Preclínica de Medicamentos , Femenino , Orden Génico , Terapia Genética/métodos , Terapia Genética/normas , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Masculino , Ratones , Mutación , Distribución Tisular , Resultado del Tratamiento
15.
Future Oncol ; 15(33): 3861-3876, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31668087

RESUMEN

Selective delivery of therapeutic agents into solid tumors has been a major challenge impeding the achievement of long-term disease remission and cure. The need to develop alternative drug delivery routes to achieve higher drug concentration in tumor tissue, reduce unwanted off-target side effects and thus achieve greater therapeutic efficacy, has resulted in an explosive body of research. Bifidobacterium spp. are anaerobic, nonpathogenic, Gram-positive bacteria, commensal to the human gut that are a possible anticancer drug-delivery vehicle. In this review, we describe Bifidobacterium's microbiology, current clinical applications, overview of the preclinical work investigating Bifidobacterium's potential to deliver anticancer therapy, and review the different strategies used up to date. Finally, we discuss both current challenges and future prospects.


Asunto(s)
Bifidobacterium/fisiología , Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Oncología Médica/métodos , Neoplasias/terapia , Medicina de Precisión/métodos , Animales , Antineoplásicos/administración & dosificación , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Genes Reporteros/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Oncología Médica/tendencias , Neoplasias/genética , Neoplasias/inmunología , Plásmidos/genética , Medicina de Precisión/tendencias , Probióticos/administración & dosificación , Resultado del Tratamiento
16.
Sci Rep ; 9(1): 11146, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31366942

RESUMEN

Targeting specific neuronal cell types is a major challenge for unraveling their function and utilizing specific cells for gene therapy strategies. Viral vector tools are widely used to target specific cells or circuits for these purposes. Here, we use viral vectors with short promoters of neuropeptide genes to target distinct neuronal populations in the hypothalamus of rats and mice. We show that lowering the amount of genomic copies is effective in increasing specificity of a melanin-concentrating hormone promoter. However, since too low titers reduce transduction efficacy, there is an optimal titer for achieving high specificity and sufficient efficacy. Other previously identified neuropeptide promoters as those for oxytocin and orexin require further sequence optimization to increase target specificity. We conclude that promoter-driven viral vectors should be used with caution in order to target cells specifically.


Asunto(s)
Vectores Genéticos/administración & dosificación , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/administración & dosificación , Regiones Promotoras Genéticas/genética , Animales , Hormonas Hipotalámicas/genética , Melaninas/genética , Ratones , Ratones Endogámicos C57BL , Orexinas/genética , Oxitocina/genética , Hormonas Hipofisarias/genética , Ratas , Ratas Long-Evans , Ratas Wistar
17.
Brain ; 142(8): 2402-2416, 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31243443

RESUMEN

Ever since its introduction 40 years ago l-3,4-dihydroxyphenylalanine (l-DOPA) therapy has retained its role as the leading standard medication for patients with Parkinson's disease. With time, however, the shortcomings of oral l-DOPA treatment have become apparent, particularly the motor fluctuations and troublesome dyskinetic side effects. These side effects, which are caused by the excessive swings in striatal dopamine caused by intermittent oral delivery, can be avoided by delivering l-DOPA in a more continuous manner. Local gene delivery of the l-DOPA synthesizing enzymes, tyrosine hydroxylase and guanosine-tri-phosphate-cyclohydrolase-1, offers a new approach to a more refined dopaminergic therapy where l-DOPA is delivered continuously at the site where it is needed i.e. the striatum. In this study we have explored the therapeutic efficacy of adeno-associated viral vector-mediated l-DOPA delivery to the putamen in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated rhesus monkeys, the standard non-human primate model of Parkinson's disease. Viral vector delivery of the two enzymes, tyrosine hydroxylase and guanosine-5'-tri-phosphate-cyclohydrolase-1, bilaterally into the dopamine-depleted putamen, induced a significant, dose-dependent improvement of motor behaviour up to a level identical to that obtained with the optimal dose of peripheral l-DOPA. Importantly, this improvement in motor function was obtained without any adverse dyskinetic effects. These results provide proof-of-principle for continuous vector-mediated l-DOPA synthesis as a novel therapeutic strategy for Parkinson's disease. The constant, local supply of l-DOPA obtained with this approach holds promise as an efficient one-time treatment that can provide long-lasting clinical improvement and at the same time prevent the appearance of motor fluctuations and dyskinetic side effects associated with standard oral dopaminergic medication.


Asunto(s)
Antiparkinsonianos/administración & dosificación , GTP Ciclohidrolasa/administración & dosificación , Vectores Genéticos/uso terapéutico , Levodopa/biosíntesis , Trastornos Parkinsonianos/terapia , Putamen/metabolismo , Tirosina 3-Monooxigenasa/administración & dosificación , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/análogos & derivados , Animales , Antiparkinsonianos/uso terapéutico , Dependovirus/genética , Evaluación Preclínica de Medicamentos , Femenino , GTP Ciclohidrolasa/análisis , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Genes Reporteros , Genes Sintéticos , Vectores Genéticos/administración & dosificación , Humanos , Macaca mulatta , Masculino , Actividad Motora/efectos de los fármacos , Trastornos Parkinsonianos/inducido químicamente , Porción Compacta de la Sustancia Negra/química , Porción Compacta de la Sustancia Negra/patología , Prueba de Estudio Conceptual , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/análisis , Proteínas Recombinantes/uso terapéutico , Tirosina 3-Monooxigenasa/análisis , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
18.
Hum Gene Ther ; 30(9): 1093-1100, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31084364

RESUMEN

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of metabolism characterized by extremely elevated total homocysteine (tHcy) in the blood. Patients diagnosed with CBS deficiency have a variety of clinical problems, including dislocated lenses, osteoporosis, cognitive and behavioral issues, and a significantly increased risk of thrombosis. Current treatment strategies involve a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine. Here, a mouse model for CBS deficiency (Tg-I278T Cbs-/-) was used to evaluate the potential of minicircle-based naked DNA gene therapy to treat CBS deficiency. A 2.3 kb DNA-minicircle containing the liver-specific P3 promoter driving the human CBS cDNA (MC.P3-hCBS) was delivered into Tg-I278T Cbs-/- mice via a single hydrodynamic tail vein injection. Mean serum tHcy decreased from 351 µM before injection to 176 µM 7 days after injection (p = 0.0005), and remained decreased for at least 42 days. Western blot analysis reveals significant minicircle-directed CBS expression in the liver tissue. Liver CBS activity increased 34-fold (12.8 vs. 432 units; p = 0.0004) in MC.P3-hCBS-injected animals. Injection of MC.P3-hCBS in young mice, subsequently followed for 202 days, showed that the vector can ameliorate the mouse homocystinuria alopecia phenotype. The present findings show that minicircle-based gene therapy can lower tHcy in a mouse model of CBS deficiency.


Asunto(s)
Cistationina betasintasa/genética , ADN Circular/genética , Terapia Genética , Vectores Genéticos/genética , Homocistinuria/genética , Homocistinuria/terapia , Animales , Biomarcadores , Cistationina betasintasa/sangre , Cistationina betasintasa/deficiencia , ADN Circular/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Homocistinuria/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Transfección/métodos , Resultado del Tratamiento
19.
J Neurosci ; 39(23): 4576-4594, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936242

RESUMEN

An innocuous sensory stimulus that reliably signals an upcoming aversive event can be conditioned to elicit locomotion to a safe location before the aversive outcome ensues. The neural circuits that mediate the expression of this signaled locomotor action, known as signaled active avoidance, have not been identified. While exploring sensorimotor midbrain circuits in mice of either sex, we found that excitation of GABAergic cells in the substantia nigra pars reticulata blocks signaled active avoidance by inhibiting cells in the pedunculopontine tegmental nucleus (PPT), not by inhibiting cells in the superior colliculus or thalamus. Direct inhibition of putative-glutamatergic PPT cells, excitation of GABAergic PPT cells, or excitation of GABAergic afferents in PPT, abolish signaled active avoidance. Conversely, excitation of putative-glutamatergic PPT cells, or inhibition of GABAergic PPT cells, can be tuned to drive avoidance responses. The PPT is an essential junction for the expression of signaled active avoidance gated by nigral and other synaptic afferents.SIGNIFICANCE STATEMENT When a harmful situation is signaled by a sensory stimulus (e.g., street light), subjects typically learn to respond with active or passive avoidance responses that circumvent the threat. During signaled active avoidance behavior, subjects move away to avoid a threat signaled by a preceding innocuous stimulus. We identified a part of the midbrain essential to process the signal and avoid the threat. Inhibition of neurons in this area eliminates avoidance responses to the signal but preserves escape responses caused by presentation of the threat. The results highlight an essential part of the neural circuits that mediate signaled active avoidance behavior.


Asunto(s)
Reacción de Prevención/fisiología , Reacción de Fuga/fisiología , Neuronas GABAérgicas/fisiología , Red Nerviosa/fisiología , Porción Reticular de la Sustancia Negra/fisiología , Núcleo Tegmental Pedunculopontino/fisiología , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/efectos de la radiación , Mapeo Encefálico , Proteínas Portadoras/genética , Proteínas Portadoras/efectos de la radiación , Clozapina/análogos & derivados , Clozapina/farmacología , Condicionamiento Clásico , Dependovirus/genética , Conducta de Ingestión de Líquido , Electrochoque , Reacción de Fuga/efectos de los fármacos , Reacción de Fuga/efectos de la radiación , Mutación con Ganancia de Función , Genes Reporteros , Vectores Genéticos/administración & dosificación , Luz , Ratones , Ruido/efectos adversos , Optogenética , Porción Reticular de la Sustancia Negra/citología , Tiempo de Reacción , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/efectos de la radiación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/efectos de la radiación , Colículos Superiores/citología , Colículos Superiores/fisiología , Tálamo/citología , Tálamo/fisiología
20.
Mol Pharm ; 16(4): 1596-1605, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30865465

RESUMEN

Glatiramer acetate (GA) is the active substance of Teva's Copaxone drug, which contains random polypeptides used to treat multiple sclerosis. Glatiramer acetate was originally developed to emulate human myelin basic protein, which contains four different residues [alanine (A), glutamic acid (E), tyrosine (T), and lysine (K)]. We found that GA can complex, condense, and transfect plasmid DNA. Mixing the positively charged GA and the negatively charged genetic material in correct proportions produced small, stable, and highly positively charged nanoparticles. This simple GA-pDNA formulation produced high levels of transfection efficiency with low toxicity in HeLa and A549 cells (lung and cervical cancer cells). Additionally, we studied and compared the nanoparticle properties, gene expression, and cytotoxicity of K100-pDNA (high-molecular-weight polylysine) and K9-pDNA (low-molecular-weight polylysine) nanoparticles to those of GA-pDNA nanoparticles. We also studied the effect of calcium, which was previously reported to reduce the size and enhance gene expression resulting from similar polyelectrolyte complexes. Adding calcium did not reduce particle size, nor improve the transfection efficiency of GA-pDNA nanoparticles as it did for polylysine-pDNA nanoparticles. GA-pDNA nanoparticles may be prepared by mixing a genetic payload with approved GA therapeutics (e.g., Copaxone), thus offering intriguing possibilities for translational gene therapy studies.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Proliferación Celular , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Acetato de Glatiramer/administración & dosificación , Nanopartículas/administración & dosificación , Polietileneimina/química , Células A549 , Células HeLa , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA