Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39315713

RESUMEN

Base editing could correct nonsense mutations that cause cystic fibrosis (CF), but clinical development is limited by the lack of delivery methods that efficiently breach the barriers presented by airway epithelia. Here, we present a novel amphiphilic shuttle peptide based on the previously reported S10 peptide that substantially improved base editor ribonucleoprotein (RNP) delivery. Studies of the S10 secondary structure revealed that the alpha-helix formed by the endosomal leakage domain (ELD), but not the cell penetrating peptide (CPP), was functionally important for delivery. By isolating and extending the ELD, we created a novel shuttle peptide, termed S237. While S237 achieved lower delivery of green fluorescent protein, it outperformed S10 at Cas9 RNP delivery to cultured human airway epithelial cells and to pig airway epithelia in vivo, possibly due to its lower net charge. In well-differentiated primary human airway epithelial cell cultures, S237 achieved a 4.6-fold increase in base editor RNP delivery, correcting up to 9.4% of the cystic fibrosis transmembrane conductance regulator (CFTR) R553X allele and restoring CFTR channel function close to non-CF levels. These findings deepen the understanding of peptide-mediated delivery and offer a translational approach for base editor RNP delivery for CF airway disease.

2.
Biomacromolecules ; 24(11): 4890-4900, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37862236

RESUMEN

The cell membrane is a restrictive biological barrier, especially for large, charged molecules, such as proteins. The use of cell-penetrating peptides (CPPs) can facilitate the delivery of proteins, protein complexes, and peptides across the membrane by a variety of mechanisms that are all limited by endosomal sequestration. To improve CPP-mediated delivery, we previously reported the rapid and effective cytosolic delivery of proteins in vitro and in vivo by their coadministration with the peptide S10, which combines a CPP and an endosomal leakage domain. Amphiphilic peptides with hydrophobic properties, such as S10, can interact with lipids to destabilize the cell membrane, thus promoting cargo internalization or escape from endosomal entrapment. However, acute membrane destabilization can result in a dose-limiting cytotoxicity. In this context, the partial or transient deactivation of S10 by modification with methoxy poly(ethylene glycol) (mPEG; i.e., PEGylation) may provide the means to alter membrane destabilization kinetics, thereby attenuating the impact of acute permeabilization on cell viability. This study investigates the influence of PEGylation parameters (molecular weight, architecture, and conjugation chemistry) on the delivery efficiency of a green fluorescent protein tagged with a nuclear localization signal (GFP-NLS) and cytotoxicity on cells in vitro. Results suggest that PEGylation mostly interferes with adsorption and secondary structure formation of S10 at the cell membrane, and this effect is exacerbated by the mPEG molecular weight. This effect can be compensated for by increasing the concentration of conjugates prepared with lower molecular weight mPEG (5 to ∼20 kDa) but not for conjugates prepared with higher molecular weight mPEG (40 kDa). For conjugates prepared with moderate-to-high molecular weight mPEG (10 to 20 kDa), partial compensation of inactivation could be achieved by the inclusion of a reducible disulfide bond, which provides a mechanism to liberate the S10 from the polymer. Grafting multiple copies of S10 to a high-molecular-weight multiarmed PEG (40 kDa) improved GFP-NLS delivery efficiency. However, these constructs were more cytotoxic than the native peptide. Considering that PEGylation could be harnessed for altering the pharmacokinetics and biodistribution profiles of peptide-based delivery agents in vivo, the trends observed herein provide new perspectives on how to manipulate the membrane permeabilization process, which is an important variable for achieving delivery.


Asunto(s)
Péptidos de Penetración Celular , Polietilenglicoles , Distribución Tisular , Polietilenglicoles/química , Polímeros/química , Péptidos de Penetración Celular/farmacología , Señales de Localización Nuclear
3.
Curr Gene Ther ; 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39225214

RESUMEN

Developing delivery vectors capable of transducing genetic material across the lung epithelia and mucus barrier is a major challenge and of great interest to enable gene therapies to treat pulmonary diseases. Recombinant Adeno-associated Viruses (rAAVs) have emerged as attractive candidates among viral and non-viral vectors due to their broad tissue tropism, ability to transduce dividing and quiescent cells, and their safety profile in current human applications. While rAAVs have demonstrated safety in earlier clinical trials for lung disease applications, there are still some limitations regarding rAAV-transgene delivery in pulmonary cells. Thus, further improvements in rAAV engineering are needed to enhance the effectiveness of rAAV-based therapies for lung diseases. Such therapies could benefit patients with chronic lung diseases, such as asthma, chronic obstructive pulmonary disease, pulmonary hypertension, and cystic fibrosis, among others, by regulating hereditary gene mutations or acquired gene deregulations causing these conditions. Alongside therapeutic development, advances in the rAAV production process are essential to meet increasing production demands, while reducing manufacturing costs. This review discusses current challenges and recent advances in the field of rAAV engineering and manufacturing to encourage the clinical development of new pulmonary gene therapy treatments.

4.
Mol Ther Nucleic Acids ; 35(3): 102290, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39233851

RESUMEN

Delivery of antisense oligonucleotides (ASOs) to airway epithelial cells is arduous due to the physiological barriers that protect the lungs and the endosomal entrapment phenomenon, which prevents ASOs from reaching their intracellular targets. Various delivery strategies involving peptide-, lipid-, and polymer-based carriers are being investigated, yet the challenge remains. S10 is a peptide-based delivery agent that enables the intracellular delivery of biomolecules such as GFP, CRISPR-associated nuclease ribonucleoprotein (RNP), base editor RNP, and a fluorescent peptide into lung cells after intranasal or intratracheal administrations to mice, ferrets, and rhesus monkeys. Herein, we demonstrate that covalently attaching S10 to a fluorescently labeled peptide or a functional splice-switching phosphorodiamidate morpholino oligomer improves their intracellular delivery to airway epithelia in mice after a single intranasal instillation. Data reveal a homogeneous delivery from the trachea to the distal region of the lungs, specifically into the cells lining the airway. Quantitative measurements further highlight that conjugation via a disulfide bond through a pegylated (PEG) linker was the most beneficial strategy compared with direct conjugation (without the PEG linker) or conjugation via a permanent thiol-maleimide bond. We believe that S10-based conjugation provides a great strategy to achieve intracellular delivery of peptides and ASOs with therapeutic properties in lungs.

5.
Front Immunol ; 14: 1231916, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37675109

RESUMEN

Introduction: Natural Killer (NK) cells hold the potential to shift cell therapy from a complex autologous option to a universal off-the-shelf one. Although NK cells have demonstrated efficacy and safety in the treatment of leukemia, the limited efficacy of NK cell-based immunotherapies against solid tumors still represents a major hurdle. In the immunosuppressive tumor microenvironment (TME), inhibitory interactions between cancer and immune cells impair antitumoral immunity. KLRC1 gene encodes the NK cell inhibitory receptor NKG2A, which is a potent NK cell immune checkpoint. NKG2A specifically binds HLA-E, a non-classical HLA class I molecule frequently overexpressed in tumors, leading to the transmission of inhibitory signals that strongly impair NK cell function. Methods: To restore NK cell cytotoxicity against HLA-E+ tumors, we have targeted the NKG2A/HLA-E immune checkpoint by using a CRISPR-mediated KLRC1 gene editing. Results: KLRC1 knockout resulted in a reduction of 81% of NKG2A+ cell frequency in ex vivo expanded human NK cells post-cell sorting. In vitro, the overexpression of HLA-E by tumor cells significantly inhibited wild-type (WT) NK cell cytotoxicity with p-values ranging from 0.0071 to 0.0473 depending on tumor cell lines. In contrast, KLRC1 KO NK cells exhibited significantly higher cytotoxicity when compared to WT NK cells against four different HLA-E+ solid tumor cell lines, with p-values ranging from<0.0001 to 0.0154. Interestingly, a proportion of 43.5% to 60.2% of NKG2A- NK cells within the edited NK cell population was sufficient to reverse at its maximum the HLA-E-mediated inhibition of NK cell cytotoxicity. The expression of the activating receptor NKG2C was increased in KLRC1 KO NK cells and contributed to the improved NK cell cytotoxicity against HLA-E+ tumors. In vivo, the adoptive transfer of human KLRC1 KO NK cells significantly delayed tumor progression and increased survival in a xenogeneic mouse model of HLA-E+ metastatic breast cancer, as compared to WT NK cells (p = 0.0015). Conclusions: Our results demonstrate that KLRC1 knockout is an effective strategy to improve NK cell antitumor activity against HLA-E+ tumors and could be applied in the development of NK cell therapy for solid tumors.


Asunto(s)
Células Asesinas Naturales , Leucemia , Humanos , Animales , Ratones , Receptores de Células Asesinas Naturales , Transporte de Proteínas , Microambiente Tumoral , Antígenos HLA-E
6.
Hum Gene Ther ; 34(15-16): 705-718, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37335046

RESUMEN

Gene editing strategies are attractive for treating genetic pulmonary diseases such as cystic fibrosis (CF). However, challenges have included the development of safe and effective vector systems for gene editing of airway epithelia and model systems to report their efficiency and durability. The domestic ferret (Mustela putorius furo) has a high degree of conservation in lung cellular anatomy with humans, and has served as an excellent model for many types of lung diseases, including CF. In this study, we evaluated the efficiency of amphiphilic shuttle peptide S10 for protein delivery and gene editing using SpCas9, and AsCas12a (Cpf1) ribonucleoproteins (RNPs). These approaches were evaluated in proliferating ferret airway basal cells, polarized airway epithelia in vitro, and lungs in vivo, by accessing the editing efficiency using reporter ferrets and measuring indels at the ferret CFTR locus. Our results demonstrate that shuttle peptides efficiently enable delivery of reporter proteins/peptides and gene editing SpCas9 or Cpf1 RNP complexes to ferret airway epithelial cells in vitro and in vivo. We measured S10 delivery efficiency of green fluorescent protein (GFP)-nuclear localization signal (NLS) protein or SpCas9 RNP into ferret airway basal cells and fully differentiated ciliated and nonciliated epithelial cells in vitro. In vitro and in vivo gene editing efficiencies were determined by Cas/LoxP-gRNA RNP-mediated conversion of a ROSA-TG Cre recombinase reporter using transgenic primary cells and ferrets. S10/Cas9 RNP was more effective, relative to S10/Cpf1 RNP at gene editing of the ROSA-TG locus. Intratracheal lung delivery of the S10 shuttle combined with GFP-NLS protein or D-Retro-Inverso (DRI)-NLS peptide demonstrated efficiencies of protein delivery that were ∼3-fold or 14-fold greater, respectively, than the efficiency of gene editing at the ROSA-TG locus using S10/Cas9/LoxP-gRNA. Cpf1 RNPs was less effective than SpCas9 at gene editing of LoxP locus. These data demonstrate the feasibility of shuttle peptide delivery of Cas RNPs to the ferret airways and the potential utility for developing ex vivo stem cell-based and in vivo gene editing therapies for genetic pulmonary diseases such as CF.


Asunto(s)
Edición Génica , Enfermedades Pulmonares , Animales , Humanos , Edición Génica/métodos , Hurones/genética , Epitelio , Péptidos/genética , Enfermedades Pulmonares/genética , Sistemas CRISPR-Cas
7.
Res Sq ; 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36824928

RESUMEN

Gene editing strategies for cystic fibrosis are challenged by the complex barrier properties of airway epithelia. We previously reported that the amphiphilic S10 shuttle peptide non-covalently combined with CRISPR-associated (Cas) ribonucleoprotein (RNP) enabled editing of human and mouse airway epithelial cells. Here, to improve base editor RNP delivery, we optimized S10 to derive the S315 peptide. Following intratracheal aerosol of Cy5-labeled peptide cargo in rhesus macaques, we confirmed delivery throughout the respiratory tract. Subsequently, we targeted CCR5 with co-administration of ABE8e-Cas9 RNP and S315. We achieved editing efficiencies of up to 5.3% in rhesus airway epithelia. Moreover, we documented persistence of edited epithelia for up to 12 months in mice. Finally, delivery of ABE8e-Cas9 targeting the CFTR R553X mutation restored anion channel function in cultured human airway epithelial cells. These results demonstrate the therapeutic potential of base editor delivery with S315 to functionally correct the CFTR R553X mutation in respiratory epithelia.

8.
Nat Commun ; 14(1): 8051, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052872

RESUMEN

Gene editing strategies for cystic fibrosis are challenged by the complex barrier properties of airway epithelia. We previously reported that the amphiphilic S10 shuttle peptide non-covalently combined with CRISPR-associated (Cas) ribonucleoprotein (RNP) enabled editing of human and mouse airway epithelial cells. Here, we derive the S315 peptide as an improvement over S10 in delivering base editor RNP. Following intratracheal aerosol delivery of Cy5-labeled peptide in rhesus macaques, we confirm delivery throughout the respiratory tract. Subsequently, we target CCR5 with co-administration of ABE8e-Cas9 RNP and S315. We achieve editing efficiencies of up-to 5.3% in rhesus airway epithelia. Moreover, we document persistence of edited epithelia for up to 12 months in mice. Finally, delivery of ABE8e-Cas9 targeting the CFTR R553X mutation restores anion channel function in cultured human airway epithelia. These results demonstrate the therapeutic potential of base editor delivery with S315 to functionally correct the CFTR R553X mutation in respiratory epithelia.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Células Epiteliales , Animales , Humanos , Ratones , Macaca mulatta/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Ribonucleoproteínas/metabolismo , Péptidos/genética , Sistemas CRISPR-Cas
9.
Biomed Eng Online ; 11: 67, 2012 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-22963645

RESUMEN

BACKGROUND: Transferring genes and drugs into cells is central to how we now study, identify and treat diseases. Several non-viral gene therapy methods that rely on the mechanical disruption of the plasma membrane have been proposed, but the success of these methods has been limited due to a lack of understanding of the mechanical parameters that lead to cell membrane permeability. METHODS: We use a simple jet of inert gas to induce local transfection of plasmid DNA both in vitro (HeLa cells) and in vivo (chicken chorioallantoic membrane). Five different capillary tube inner diameters and three different gases were used to treat the cells to understand the dependency of transfection efficiency on the dynamic parameters. RESULTS: The simple setup has the advantage of allowing us to calculate the forces acting on cells during transfection. We found permeabilization efficiency was related to the dynamic pressure of the jet. The range of dynamic pressures that led to transfection in HeLa cells was small (200 ± 20 Pa) above which cell stripping occurred. We determined that the temporary pores allow the passage of dextran up to 40 kDa and reclose in less than 5 seconds after treatment. The optimized parameters were also successfully tested in vivo using the chorioallantoic membrane of the chick embryo. CONCLUSIONS: The results show that the number of cells transfected with the plasmid scales with the dynamic pressure of the jet. Our results show that mechanical methods have a very small window in which cells are permeabilized without injury (200 to 290 Pa). This simple apparatus helps define the forces needed for physical cell transfection methods.


Asunto(s)
Fenómenos Mecánicos , Gases Nobles , Transfección/instrumentación , Animales , Adhesión Celular , Muerte Celular , Permeabilidad de la Membrana Celular , Membrana Corioalantoides/metabolismo , Vidrio , Células HeLa , Humanos
10.
Consult Pharm ; 27(12): 857-67, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23229074

RESUMEN

OBJECTIVE: Vilazodone (VIIBRYD, Trovis Pharmaceuticals; New Haven, Connecticut, also known as 659746, EMD68843, SB-659746-A) is a newly introduced antidepressant that has taken approximately a decade from its discovery to approval by the Food and Drug Administration. This paper will review the chemistry, pharmacodynamics, pharmacokinetics, clinical efficacy, tolerability, drug-drug interaction potential, dosing, and administration of this agent. DATA SOURCES: Medline/PubMed/IPA/EMBASE databases were searched using the terms "vilazodone," "659746," "EMD68843," and "SB-659746-A." All English-language papers from 1985 to April 2012 were reviewed for relevance. Bibliographies of all papers were reviewed to identify further papers. STUDY SELECTION: All English-language papers from 1985 to present appearing in these searches were reviewed for relevance to this paper. In addition, their bibliographies were reviewed to identify any papers not identified in the searches. Data are expressed as mean or mean ± standard deviation, unless otherwise noted. DATA SYNTHESIS: Vilazodone is the first combined selective serotonin reuptake inhibitor (SSRI)/5-HT1A receptor agonist antidepressant. Vilazodone must be administered with food to optimize bioavailability. The primary route of elimination is metabolism followed by excretion of metabolites. Advancing age and renal and hepatic impairment do not alter its disposition. Early phase II clinical trials were unable to demonstrate antidepressant efficacy. However, later phase III trials using 40 mg daily doses were able to demonstrate superior efficacy compared with placebo treatment. Adverse events, warnings, and precautions mirror those of other SSRIs. CONCLUSION: Although there are theoretical reasons why 5-HT1A agonism may be a desirable additional property in antidepressants, there is no evidence to date that vilazodone has any advantage over existing post-tricyclic antidepressants. It has a narrow therapeutic dosing range whose upper boundary is close to that producing intolerable gastrointestinal and central nervous system adverse events. Further research will clarify and refine the role of vilazodone in the management of psychiatric disorders.


Asunto(s)
Antidepresivos/uso terapéutico , Benzofuranos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Indoles/uso terapéutico , Piperazinas/uso terapéutico , Animales , Antidepresivos/efectos adversos , Antidepresivos/farmacología , Benzofuranos/efectos adversos , Benzofuranos/farmacología , Disponibilidad Biológica , Ensayos Clínicos como Asunto , Trastorno Depresivo Mayor/fisiopatología , Interacciones Farmacológicas , Humanos , Indoles/efectos adversos , Indoles/farmacología , Piperazinas/efectos adversos , Piperazinas/farmacología , Agonistas del Receptor de Serotonina 5-HT1/efectos adversos , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Agonistas del Receptor de Serotonina 5-HT1/uso terapéutico , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Clorhidrato de Vilazodona
11.
Mol Cancer ; 10: 145, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-22118625

RESUMEN

BACKGROUND: YB-1 is a multifunctional protein that affects transcription, splicing, and translation. Overexpression of YB-1 in breast cancers causes cisplatin resistance. Recent data have shown that YB-1 is also overexpress in colorectal cancer. In this study, we tested the hypothesis that YB-1 also confers oxaliplatin resistance in colorectal adenocarcinomas. RESULTS: We show for the first time that transfection of YB-1 cDNA confers oxaliplatin resistance in two colorectal cancer cell lines (SW480 and HT29 cell lines). Furthermore, we identified by mass spectrometry analyses important YB-1 interactors required for such oxaliplatin resistance in these colorectal cancer cell lines. A tagged YB-1 construct was used to identify proteins interacting directly to YB-1 in such cells. We then focused on proteins that are potentially involved in colorectal cancer progression based on the Oncomine microarray database. Genes encoding for these YB-1 interactors were also examined in the public NCBI comparative genomic hybridization database to determine whether these genes are localized to regions of chromosomes rearranged in colorectal cancer tissues. From these analyses, we obtained a list of proteins interacting with YB-1 and potentially involved in oxaliplatin resistance. Oxaliplatin dose response curves of SW480 and HT29 colorectal cancer cell lines transfected with several siRNAs corresponding to each of these YB-1 interactors were obtained to identify proteins significantly affecting oxaliplatin sensitivity upon gene silencing. Only the depletion of either NONO or RALY sensitized both colorectal cancer cell lines to oxaliplatin. Furthermore, depletion of NONO or RALY sensitized otherwise oxaliplatin resistant overexpressing YB-1 SW480 or HT29 cells. CONCLUSION: These results suggest knocking down NONO or RALY significant counteracts oxaliplatin resistance in colorectal cancers overexpressing the YB-1 protein.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Ribonucleoproteína Heterogénea-Nuclear Grupo C/genética , Proteínas Asociadas a Matriz Nuclear/genética , Factores de Transcripción de Octámeros/genética , Compuestos Organoplatinos/farmacología , Proteínas de Unión al ARN/genética , Proteína 1 de Unión a la Caja Y/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Proteínas de Unión al ADN , Resistencia a Antineoplásicos , Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , Humanos , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Proteínas de Unión al ARN/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo
12.
Cancer Sci ; 102(7): 1410-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21466612

RESUMEN

The Y-box binding protein 1 (YB-1) is a multifunctional protein that affects transcription, splicing, and translation. Overexpression of YB-1 in breast cancers causes cisplatin resistance. The exact mechanism by which YB-1 confers cisplatin resistance is unknown. The aim of the present study was to identify, using mass spectrometry, proteins that interact with YB-1 that are important for cisplatin resistance in two breast cancer cell lines, namely MCF7 and MDA-MB-231. A tagged YB-1 construct was used to identify proteins interacting directly with YB-1 in breast cancer cells. We then focused on proteins that are potentially involved in breast cancer progression based on the ONCOMINE public microarray database. Genes encoding for these YB-1-interacting proteins were examined in the public NCBI comparative genomic hybridization database to determine whether they are localized to regions of chromosomes that are rearranged in breast cancer tissues. From these analyses, we generated a list of proteins potentially involved in cisplatin resistance. Cisplatin dose-response curves were constructed in MCF7 and MDA-MB-231 transfected with four siRNA corresponding to each of these YB-1 interactors to identify proteins significantly affecting cisplatin sensitivity upon gene silencing. Depletion of only the X-linked ribosomal protein S4 (RPS4X) resulted in consistent resistance to cisplatin in both cell lines with at least three different siRNA sequences against RPS4X. Further analyses indicated that the knock down of RPS4X decreased DNA synthesis, induced cisplatin resistance, and is equivalent to the overexpression of YB-1 in both MCF7 and MDA-MB-231 cells. These results suggest that the RPS4X/YB-1 complex is a significant potential target to counteract cisplatin resistance in breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Cisplatino/farmacología , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Proteínas Ribosómicas/fisiología , Neoplasias de la Mama/patología , Bromodesoxiuridina/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/análisis , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Femenino , Humanos , Proteínas Nucleares/análisis , ARN Interferente Pequeño/genética , Proteínas Ribosómicas/análisis , Proteína 1 de Unión a la Caja Y
13.
DNA Repair (Amst) ; 8(6): 704-19, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19272840

RESUMEN

Maintenance of the mitochondrial genome (mtDNA) is essential for proper cellular function. The accumulation of damage and mutations in the mtDNA leads to diseases, cancer, and aging. Mammalian mitochondria have proficient base excision repair, but the existence of other DNA repair pathways is still unclear. Deficiencies in DNA mismatch repair (MMR), which corrects base mismatches and small loops, are associated with DNA microsatellite instability, accumulation of mutations, and cancer. MMR proteins have been identified in yeast and coral mitochondria; however, MMR proteins and function have not yet been detected in human mitochondria. Here we show that human mitochondria have a robust mismatch-repair activity, which is distinct from nuclear MMR. Key nuclear MMR factors were not detected in mitochondria, and similar mismatch-binding activity was observed in mitochondrial extracts from cells lacking MSH2, suggesting distinctive pathways for nuclear and mitochondrial MMR. We identified the repair factor YB-1 as a key candidate for a mitochondrial mismatch-binding protein. This protein localizes to mitochondria in human cells, and contributes significantly to the mismatch-binding and mismatch-repair activity detected in HeLa mitochondrial extracts, which are significantly decreased when the intracellular levels of YB-1 are diminished. Moreover, YB-1 depletion in cells increases mitochondrial DNA mutagenesis. Our results show that human mitochondria contain a functional MMR repair pathway in which YB-1 participates, likely in the mismatch-binding and recognition steps.


Asunto(s)
Reparación de la Incompatibilidad de ADN , ADN Mitocondrial/genética , Proteínas de Unión al ADN/metabolismo , Mitocondrias/metabolismo , Proteínas Nucleares/metabolismo , Núcleo Celular/metabolismo , Resistencia al Cloranfenicol , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Células HeLa , Humanos , Proteínas Nucleares/genética , Consumo de Oxígeno , Fracciones Subcelulares , Proteína 1 de Unión a la Caja Y
14.
Consult Pharm ; 25(12): 816-28, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21172762

RESUMEN

OBJECTIVE: To review the chemistry, pharmacodynamics, pharmacokinetics, efficacy, tolerability, dosing, and role of the Osmotic-controlled Release Oral delivery System (OROS) hydromorphone extended-release (ER) tablets. DATA SOURCE: A MEDLINE/PUBMED search (1986-August 2010) was conducted to identify studies in the English language, with additional references being obtained from their bibliographies. STUDY SELECTION: All studies of hydromorphone ER were reviewed. DATA SYNTHESIS: This is the second long-acting hydromorphone formulation to receive approval by the Food and Drug Administration (a twice-daily formulation was approved in September 2004, but was subsequently withdrawn in July 2005). Hydromorphone is a semi-synthetic mu-opioid receptor agonist structurally similar to morphine, hydrocodone, and oxymorphone. OROS ER technology allows once-daily dosing. Clinical trials have focused on the convertibility of (an) other opioid(s) to hydromorphone ER in chronic malignant and nonmalignant pain. This product displays the expected opioid side effects, being comparable to oxycodone controlled-release. Coadministration with ethanol does not produce the degree of "dose-dumping" seen with the former hydromorphone twice-daily product or oxymorphone ER. Hydromorphone ER is indicated for the management of moderate-to-severe pain in opioidtolerant patients requiring continuous, around-the-clock opioid analgesia for an extended period of time. Dosage adjustment is recommended in patients with moderate hepatic impairment (Child-Pugh class B) and moderate renal impairment (creatinine clearance of 30-60 mL/min). CONCLUSION: Hydromorphone ER is the newest oral opioid to enter a crowded marketplace now totaling 15 different Schedule 2 opioids (including tapentadol), and tramadol, available in oral, parenteral, rectal, transdermal, transmucosal, and intranasal formulations. It does not appear to have any unique assets or liabilities and should be considered as one of many oral opioids available for the management of persistent pain of moderate-to-severe intensity.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Hidromorfona/administración & dosificación , Dolor/tratamiento farmacológico , Administración Oral , Anciano , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Preparaciones de Acción Retardada , Esquema de Medicación , Humanos , Hidromorfona/efectos adversos , Hidromorfona/farmacología , Presión Osmótica
15.
Drugs ; 69(7): 775-807, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19441868

RESUMEN

Urinary tract infection (UTI) refers to the presence of clinical signs and symptoms arising from the genitourinary tract plus the presence of one or more micro-organisms in the urine exceeding a threshold value for significance (ranges from 102 to 103 colony-forming units/mL). Infections are localized to the bladder (cystitis), renal parenchyma (pyelonephritis) or prostate (acute or chronic bacterial prostatitis). Single UTI episodes are very common, especially in adult women where there is a 50-fold predominance compared with adult men. In addition, recurrent UTIs are also common, occurring in up to one-third of women after first-episode UTIs. Recurrences requiring intervention are usually defined as two or more episodes over 6 months or three or more episodes over 1 year (this definition applies only to young women with acute uncomplicated UTIs). A cornerstone of prevention of UTI recurrence has been the use of low-dose once-daily or post-coital antimicrobials; however, much interest has surrounded non-antimicrobial-based approaches undergoing investigation such as use of probiotics, vaccines, oligosaccharide inhibitors of bacterial adherence and colonization, and bacterial interference with immunoreactive extracts of Escherichia coli. Local (intravaginal) estrogen therapy has had mixed results to date. Cranberry products in a variety of formulations have also undergone extensive evaluation over several decades in the management of UTIs. At present, there is no evidence that cranberry can be used to treat UTIs. Hence, the focus has been on its use as a preventative strategy. Cranberry has been effective in vitro and in vivo in animals for the prevention of UTI. Cranberry appears to work by inhibiting the adhesion of type I and P-fimbriated uropathogens (e.g. uropathogenic E. coli) to the uroepithelium, thus impairing colonization and subsequent infection. The isolation of the component(s) of cranberry with this activity has been a daunting task, considering the hundreds of compounds found in the fruit and its juice derivatives. Reasonable evidence suggests that the anthocyanidin/proanthocyanidin moieties are potent antiadhesion compounds. However, problems still exist with standardization of cranberry products, which makes it extremely difficult to compare products or extrapolate results. Unfortunately, most clinical trials have had design deficiencies and none have evaluated specific key cranberry-derived compounds considered likely to be active moieties (e.g. proanthocyanidins). In general, the preventive efficacy of cranberry has been variable and modest at best. Meta-analyses have established that recurrence rates over 1 year are reduced approximately 35% in young to middle-aged women. The efficacy of cranberry in other groups (i.e. elderly, paediatric patients, those with neurogenic bladder, those with chronic indwelling urinary catheters) is questionable. Withdrawal rates have been quite high (up to 55%), suggesting that these products may not be acceptable over long periods. Adverse events include gastrointestinal intolerance, weight gain (due to the excessive calorie load) and drug-cranberry interactions (due to the inhibitory effect of flavonoids on cytochrome P450-mediated drug metabolism). The findings of the Cochrane Collaboration support the potential use of cranberry products in the prophylaxis of recurrent UTIs in young and middle-aged women. However, in light of the heterogeneity of clinical study designs and the lack of consensus regarding the dosage regimen and formulation to use, cranberry products cannot be recommended for the prophylaxis of recurrent UTIs at this time.


Asunto(s)
Bebidas , Infecciones Urinarias/tratamiento farmacológico , Vaccinium macrocarpon/química , Adulto , Animales , Niño , Ensayos Clínicos como Asunto , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Fitoterapia
16.
Clin Ther ; 31(1): 1-31, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19243704

RESUMEN

BACKGROUND: Paraphilias are characterized by recurrent, intense, sexually arousing fantasies, urges, or behaviors, over a period of > or =6 months, generally involving nonhuman objects, suffering or humiliation of oneself or one's partner, or children or other nonconsenting persons. These fantasies, urges, and behaviors produce clinically significant distress or impairments in social, occupational, and other important areas of functioning. OBJECTIVE: The goal of this article was to provide an in-depth review of the clinical pharmacology of the main antiandrogens (cyproterone acetate, medroxyprogesterone acetate [MPA], and the luteinizing hormone-releasing hormone [LHRH] agonists) used in the treatment of the paraphilias, as well as a discussion of the relevant clinical case reports, case series, and controlled trials. Treatment recommendations are also provided. METHODS: Relevant publications were identified through a search of the English-language literature indexed on MEDLINE/PubMed (1966-September 2008) using the search terms paraphilia, sex offender, hypersexuality, sexual behaviors, fetish, transvestic fetishism, sexual addiction, sexual compulsivism, selective serotonin reuptake inhibitors, tricyclic antidepressants, antiandrogens, cyproterone acetate, medroxyprogesterone acetate, LHRH agonists, and estrogens. Additional publications were identified from the bibliographies of retrieved publications. RESULTS: In vitro and in vivo (animal) studies have revealed that serotonin and prolactin inhibit sexual arousal, while norepinephrine (via alpha(1)-adrenoceptor activation), dopamine, acetylcholine (via muscarinic receptor activation), enkephalins, oxytocin, gonadotropin-releasing hormone, follicle-stimulating hormone, luteinizing hormone, testosterone/dihydrotestosterone, and estrogen/progesterone stimulate it. Most of the currently used pharmacologic treatments of the paraphilias have serotonin and testosterone/dihydrotestosterone as their targets. Cognitive-behavioral psychotherapy should be initiated in all offenders. In those at the highest risk of reoffending, psychotherapy should be initiated at the same time as drug therapy because their combination is associated with better results compared with either as monotherapy (especially in pedophiles). In offenders committing non-"hands-on" or violent paraphilias and those at low risk of reoffending, serotoninergic monotherapies (selective serotonin reuptake inhibitors [SSRIs] or tricyclic antidepressants) are reasonable choices (SSRIs are preferred). In other offenders, initial dual combination therapy (serotoninergic plus antiandrogenic) is recommended. Progestogens should be used before LHRH agonists or estrogens. Cyproterone acetate and MPA are preferred as oral and IM progestogens, respectively. Failure of dual combination serotoninergic/ progestogen therapy should prompt a change in one or both of the components (eg, SSRI to tricyclic antidepressants or vice versa, or cyproterone acetate to MPA or vice versa) or the addition or substitution of an LHRH agonist (leuprolide or triptorelin) for the progestogen. Estrogens are second- or third-line agents. Rarely, triple combination therapy is necessary (serotoninergic plus LHRH agonist or progestogen plus estrogen). It appears that recidivism rates are reduced by the use of psychotherapy alone, drug therapy alone, and more so by their combination. CONCLUSIONS: Although some progress has been made in the therapy of paraphilic and nonparaphilic sexual disorders, much work remains to be done. The development of more specific, more effective, and better-tolerated medications for these disorders should be recognized as a program worthy of greater support from government and pharmaceutical industry sources. Clinical studies performed to date have largely been of poor design, making the recommendations provided in this review tentative at best.


Asunto(s)
Trastornos Parafílicos/tratamiento farmacológico , Delitos Sexuales/prevención & control , Disfunciones Sexuales Psicológicas/tratamiento farmacológico , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Femenino , Hormona Liberadora de Gonadotropina/agonistas , Humanos , Masculino
17.
Consult Pharm ; 24(11): 833-40, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20092221

RESUMEN

OBJECTIVE: To review the pharmacodynamics, pharmacokinetics, clinical efficacy, tolerability, dosing, and administration of tapentadol, a combination mu-opioid-receptor agonist and monoamine-reuptake inhibitor, and compare it with tramadol, the first drug in this class. DATA SOURCES: MEDLINE/PUBMED and EMBASE searches (1986 through March 2009) were conducted to identify pertinent English-language papers. In addition, meeting abstracts from multiple pain specialty organizations were searched (2000 through 2008). STUDY SELECTION AND DATA EXTRACTION: All papers/abstracts evaluating any aspect of tapentadol. DATA SYNTHESIS: Oral tapentadol hydrochloride (HCl) is the second combination mu-opioid-receptor agonist and monoamine-reuptake inhibitor to be approved by the Food and Drug Administration (approved for treatment of moderate-to-severe acute pain in adults). It was active in a wide variety of pain states in animals and humans. It behaved in a similar fashion to morphine and hydromorphone in animal and human models of physical/psychological dependence. Oral tapentadol HCI is administered in doses of 50 to 100 mg every four to six hours (dose and dosing interval being selected on the basis of pain intensity). No specific recommendations have been made for elders. CONCLUSION: Tapentadol overcomes some of the liabilities of tramadol. However, it still has some liabilities: its potential to contribute to/precipitate serotonin syndrome and anticholinergic/5-HT3 antagonist effects and to induce physical/psychological dependence (similar to schedule II opioids). There is also a dearth of information in terms of efficacy/tolerability in chronic pain states, clinical data in frail elders, and details of drug-drug interaction potential vis-à-vis glucuronidation and quantitation of the risk of serotonin syndrome.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Fenoles/uso terapéutico , Tramadol/uso terapéutico , Inhibidores de Captación Adrenérgica/efectos adversos , Inhibidores de Captación Adrenérgica/farmacología , Inhibidores de Captación Adrenérgica/uso terapéutico , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Ensayos Clínicos como Asunto , Humanos , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Fenoles/efectos adversos , Fenoles/farmacología , Receptores Opioides mu/agonistas , Tapentadol , Tramadol/efectos adversos , Tramadol/farmacología
18.
Consult Pharm ; 24(3): 210-26, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19555136

RESUMEN

OBJECTIVE: To review the chemistry, pharmacodynamics, pharmacokinetics, clinical efficacy, tolerability, indications, and dosing and administration of methylnaltrexone methobromide, the first approved peripherally selective opioid receptor-antagonist. DATA SOURCE: MEDLINE/PUBMED and EMBASE searches (January 1966-February 2009) were conducted to identify pertinent English-language studies. STUDY SELECTION AND DATA EXTRACTION: All studies evaluating any aspect of methylnaltrexone methobromide. DATA SYNTHESIS: Subcutaneous methylnaltrexone methobromide is the first opioid receptor-antagonist to be approved for the treatment of opioid receptor-agonist-induced constipation (subset with advanced disease, receiving palliative care, with an inadequate response to laxative therapy). This agent lacks meaningful activity in the central nervous system and, hence, will not compromise centrally mediated analgesia or precipitate centrally mediated signs/symptoms of opioid receptor-agonist withdrawal. There are no published comparative trials with traditional pharmacologic/nonpharmacologic laxation regimens. CONCLUSION: Methylnaltrexone methobromide is administered into the upper arm, abdomen, or thigh once every other day, with the frequency of dosing being increased, if needed, to a maximum of once daily. Recommended doses are 8 mg, 12 mg, or 0.15 mg/kg, depending on patient weight. For creatinine clearances less than 30 mL/min, the dose should be reduced by 50%. The average wholesale price is $83.33 for a 12 mg single-use vial (Medispan, accessed December 4, 2008). Clearly, parenteral agents are not as useful as oral agents and results of ongoing studies with new oral formulations of this product are eagerly awaited.


Asunto(s)
Naltrexona/análogos & derivados , Antagonistas de Narcóticos/farmacología , Animales , Ensayos Clínicos como Asunto , Interacciones Farmacológicas , Humanos , Naltrexona/efectos adversos , Naltrexona/farmacocinética , Naltrexona/farmacología , Naltrexona/uso terapéutico , Compuestos de Amonio Cuaternario/efectos adversos , Compuestos de Amonio Cuaternario/farmacocinética , Compuestos de Amonio Cuaternario/farmacología , Compuestos de Amonio Cuaternario/uso terapéutico
19.
Nat Commun ; 10(1): 4906, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31659165

RESUMEN

The delivery of biologic cargoes to airway epithelial cells is challenging due to the formidable barriers imposed by its specialized and differentiated cells. Among cargoes, recombinant proteins offer therapeutic promise but the lack of effective delivery methods limits their development. Here, we achieve protein and SpCas9 or AsCas12a ribonucleoprotein (RNP) delivery to cultured human well-differentiated airway epithelial cells and mouse lungs with engineered amphiphilic peptides. These shuttle peptides, non-covalently combined with GFP protein or CRISPR-associated nuclease (Cas) RNP, allow rapid entry into cultured human ciliated and non-ciliated epithelial cells and mouse airway epithelia. Instillation of shuttle peptides combined with SpCas9 or AsCas12a RNP achieves editing of loxP sites in airway epithelia of ROSAmT/mG mice. We observe no evidence of short-term toxicity with a widespread distribution restricted to the respiratory tract. This peptide-based technology advances potential therapeutic avenues for protein and Cas RNP delivery to refractory airway epithelial cells.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Endonucleasas/metabolismo , Células Epiteliales/metabolismo , Enfermedades Pulmonares/terapia , Pulmón/metabolismo , Péptidos/genética , Animales , Proteínas Bacterianas/genética , Bronquios/citología , Bronquios/metabolismo , Endonucleasas/genética , Terapia Genética , Humanos , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Ratones , Péptidos/administración & dosificación , Péptidos/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Porcinos
20.
Int J Biochem Cell Biol ; 40(11): 2492-507, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18571458

RESUMEN

YB-1 is a protein involved in DNA repair, transcription, splicing, translation, and confers cisplatin resistance in several cancers. However, it is unknown which YB-1 activity is required for this resistance. To identify the mechanism(s) by which nuclear YB-1 confers cisplatin resistance, we generated several YB-1 mutants and tested their impact on resistance in the mammary tumor cell lines MCF7 and MDA-MB-231. Transfection of wild type YB-1 bestowed cisplatin resistance in such cells but a mutant YB-1 with a point mutation at position 175 (YB-1(E175A)) did not. A truncated YB-1(1-205) increased cisplatin resistance above the levels conferred by wild type YB-1. The truncated YB-1(1-205) has intact nuclease activities but could not separate a DNA duplex containing a Y-box sequence (activities associated with DNA repair). Moreover, this truncated YB-1(1-205) did not alter splicing of the adenovirus E1A pre-mRNA minigene as it had low binding affinity for several splicing factors. In contrast, the mutant YB-1(E175A) protein behaved like wild type YB-1 regarding all these activities but yet did not confer cisplatin resistance. Finally, transfection of mutant YB-1(E175A) had low impact on overall transcription. The wild type and truncated YB-1(1-205) induced important but different alterations in gene expression as revealed by microarray analyses. Our results indicate that the splicing and the nuclease activities associated with YB-1 have minor impact on cisplatin resistance. In contrast, the global expression profiles displayed by both wild type and truncated YB-1(1-205) revealed several chemoresistance signatures which differed depending on the genetic status of the breast cancer cell line used.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama , Cisplatino/uso terapéutico , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Proteínas Nucleares/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Ciclo Celular/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Mutación Puntual , Empalme del ARN , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína 1 de Unión a la Caja Y
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA