Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Lang Commun Disord ; 58(2): 542-554, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36239167

RESUMEN

BACKGROUND: Primary progressive aphasia (PPA) is a rare neurodegenerative brain disorder characterized by declining language ability. There is currently no way to reverse or slow the course of the progressive brain degeneration, nor is there a cure for PPA. Throughout the course of the disease, any treatment must therefore be palliative in nature and should be designed to manage symptoms and improve the quality of life of the affected person. There is little information in the medical literature about strategies to make meaningful improvements to the quality of life of people with PPA written from the perspective of those living with this condition. AIMS: I have a clinical diagnosis of the nonfluent/agrammatic variant of PPA (nfvPPA), supported by imaging. In this report I discuss my experience of the progressive loss of language and communication skills, and detail the challenges I have been facing. I also describe how my quality of life has been enhanced by the early initiation of treatment focusing on communication strategies targeted to my specific impairments and designed to support my individual interests and goals. METHODS & PROCEDURES: I was fortunate to obtain an early diagnosis from a cognitive neurologist experienced with PPA. From the onset of my language difficulties, I have received excellent personalized care from a multidisciplinary medical team including speech-language pathologists, a cognitive neurologist and other doctors. MAIN CONTRIBUTIONS: My life during the early stage of nfvPPA has been enriched by personalized care focused on supporting the particular activities, interests and goals that are most important and meaningful to me. As my disease has progressed, I have benefited from an evolving range of strategies and adaptations targeted to the specific deficits in the areas of speaking, writing and reading that I have been facing at any given time. In addition, I have adopted methods to enhance the benefit of these language-directed strategies. And I have been employing evidence-based approaches that improve general brain health and thereby indirectly support my language. CONCLUSIONS & IMPLICATIONS: My experience represents a model for the personalized care of people in the early stage of nfvPPA. WHAT THIS PAPER ADDS: What is already known on the subject There is minimal information in the medical literature describing the subjective experience of a person with PPA. There is little information in the medical literature about strategies to make meaningful improvements to the quality of life of people in the early stage of PPA. What this paper adds to existing knowledge I have a clinical diagnosis of nfvPPA, supported by imaging. In this paper I give a first-person account of my experience of the progressive loss of language and communication skills, and I detail the challenges I have been facing. I describe how my quality of life during the early stage of nfvPPA has been enhanced by an evolving range of strategies and adaptations tailored to my speech and language deficits as they have arisen. These compensatory strategies have focused on supporting the particular activities, interests and goals that are most important and meaningful to me. What are the potential or actual clinical implications of this work? The description of my subjective experience of the progressive loss of language and communication skills offers insight for speech-language pathologists, neurologists and other professionals involved in the clinical care of people in the early stage of nfvPPA. My experience represents a model for the personalized clinical care of people in the early stage of this disorder.


Asunto(s)
Afasia Progresiva Primaria , Enfermedades Neurodegenerativas , Humanos , Afasia Progresiva Primaria/diagnóstico , Afasia Progresiva Primaria/terapia , Calidad de Vida , Encéfalo
2.
Dementia (London) ; : 14713012241281006, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39214153

RESUMEN

Primary progressive aphasia (PPA) is a neurodegenerative brain disorder characterized by declining language ability. It is a rare, often young-onset dementia with a devastating impact on the work and personal activities of those affected. At present there is no cure or disease-modifying therapy for PPA nor any way to arrest or slow the underlying progressive brain degeneration. Throughout the course of the condition any treatment must therefore be palliative-designed to manage symptoms and improve the quality of life of the affected person. The majority of those affected receive little or no follow-up care after diagnosis, particularly in the early stage of the disease. There is very little information in the medical literature about person-centered care designed to improve the quality of life of people with PPA written from the perspective of those living with this condition. I received an early and accurate clinical diagnosis of the nonfluent/agrammatic variant of PPA, supported by imaging. I am fortunate to have benefited from exemplary individualized care from a multidisciplinary medical team from the onset of my difficulties with language. In this paper, I discuss my lived experience of all aspects of this personalized and person-centered care, describing how it was founded on shared decision-making and a holistic, dementia-inclusive approach encompassing the physical, mental, emotional, psychosocial and spiritual dimensions of living with an incurable neurodegenerative disease.

3.
Lab Invest ; 93(3): 268-78, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23358109

RESUMEN

Metastatic involvement of the skeleton is a frequent consequence of advanced prostate cancer. These skeletal metastases cause a number of debilitating complications and are refractory to current treatments. New therapeutic options are being explored, including conditionally replicating adenoviruses (CRAds). CRAds are engineered to selectively replicate in and destroy tumor cells and can be 'armed' with exogenous transgenes for enhanced potency. We hypothesized that a CRAd armed with osteoprotegerin (OPG), an inhibitor of osteoclastogenesis, would inhibit the progression of prostate cancer bone metastases by directly lysing tumor cells and by reducing osteoclast activity. Although prostate cancer bone metastases are predominantly osteoblastic in nature, increased osteoclast activity is critical for the growth of these lesions. Ad5-Δ24-sOPG-Fc-RGD is a CRAd that carries a fusion of the ligand-binding domains of OPG and the Fc region of human IgG1 in place of the viral E3B genes. To circumvent low tumor cell expression of the native adenoviral receptor, an arginine-glycine-aspartic acid (RGD) peptide insertion within the viral fiber knob allows infection of cells expressing α(v) integrins. A 24-base pair deletion (Δ24) within viral E1A limits replication to cells with aberrant retinoblastoma cell cycle regulator/tumor suppressor expression. We have confirmed that Ad5-Δ24-sOPG-Fc-RGD replicates within and destroys prostate cancer cells and, in both murine and human coculture models, that infection of prostate cancer cells inhibits osteoclastogenesis in vitro. In a murine model, progression of advanced prostate cancer bone metastases was inhibited by treatment with Ad5-Δ24-sOPG-Fc-RGD but not by an unarmed control CRAd.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias Óseas/terapia , Viroterapia Oncolítica/métodos , Osteoprotegerina/farmacología , Neoplasias de la Próstata/patología , Adenoviridae/genética , Análisis de Varianza , Animales , Línea Celular Tumoral , Humanos , Inmunoglobulina G/genética , Luciferasas , Masculino , Ratones , Oligopéptidos/genética , Oligopéptidos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Microtomografía por Rayos X
4.
Mol Ther ; 20(12): 2222-33, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22948673

RESUMEN

Targeting the tumor stroma in addition to the malignant cell compartment is of paramount importance to achieve complete tumor regression. In this work, we modified a previously designed tumor stroma-targeted conditionally replicative adenovirus (CRAd) based on the SPARC promoter by introducing a mutated E1A unable to bind pRB and pseudotyped with a chimeric Ad5/3 fiber (Ad F512v1), and assessed its replication/lytic capacity in ovary cancer in vitro and in vivo. AdF512v1 was able to replicate in fresh samples obtained from patients: (i) with primary human ovary cancer; (ii) that underwent neoadjuvant treatment; (iii) with metastatic disease. In addition, we show that four intraperitoneal (i.p.) injections of 5 × 10(10) v.p. eliminated 50% of xenografted human ovary tumors disseminated in nude mice. Moreover, AdF512v1 replication in tumor models was enhanced 15-40-fold when the tumor contained a mix of malignant and SPARC-expressing stromal cells (fibroblasts and endothelial cells). Contrary to the wild-type virus, AdF512v1 was unable to replicate in normal human ovary samples while the wild-type virus can replicate. This study provides evidence on the lytic capacity of this CRAd and highlights the importance of targeting the stromal tissue in addition to the malignant cell compartment to achieve tumor regression.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Neoplasias Ováricas/genética , Células del Estroma/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Palliat Care ; 38(3): 276-281, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34210214

RESUMEN

Frontotemporal disorders are a group of rare young-onset dementias for which there is no cure, nor is there any way to slow the underlying progressive brain degeneration. To date those affected have typically received very little, if any, follow-up care after diagnosis, particularly in the early stages of their disease. I have received a clinical diagnosis, supported by imaging, of primary progressive aphasia, a form of frontotemporal degeneration characterized in the initial phase by progressive impairment of language ability. From the onset, I have been fortunate to receive excellent ongoing palliative care from a multidisciplinary team, some of whom had never previously seen anyone with this disorder. My quality of life has been enhanced by an evolving range of creative strategies and adaptations targeted to my deficits as they have arisen. In this paper, I discuss my experience of the process underlying this personalized plan, which serves as a paradigm for the development of novel palliative care approaches for people living with rare disorders, both neurodegenerative diseases and other conditions.


Asunto(s)
Afasia Progresiva Primaria , Cuidados Paliativos , Humanos , Calidad de Vida , Afasia Progresiva Primaria/terapia
6.
Dementia (London) ; 20(3): 936-951, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32276559

RESUMEN

Primary progressive aphasia is a major clinical presentation of frontotemporal lobar degeneration and is a young-onset disorder characterized by deteriorating language skills. There is currently no cure for primary progressive aphasia, nor is it possible to slow the course of the underlying progressive brain degeneration. Hence the chief goal of treatment is palliative. Although the inability to employ language at one's previous level represents a significant functional impairment for those affected, there is a dearth of information about how to make meaningful improvements to the quality of life of people in the early stages of primary progressive aphasia. I have a clinical diagnosis, supported by imaging, of the nonfluent/agrammatic variant of primary progressive aphasia and am under the care of a multidisciplinary medical team. This report is based on my ongoing experience and describes the development and implementation of an evolving set of targeted strategies and adaptations designed to enhance the quality of life of a person in the early stages of this disorder.


Asunto(s)
Adaptación Psicológica , Afasia Progresiva Primaria no Fluente , Calidad de Vida , Demencia Frontotemporal , Humanos , Estudios Longitudinales
7.
J Spinal Disord Tech ; 23(1): 63-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20084034

RESUMEN

STUDY DESIGN: Ex vivo gene transfer for spinal fusion. OBJECTIVE: This study aimed to evaluate ex vivo transfer of the nuclear-localized Hoxc-8-interacting domain of Smad1 (termed Smad1C) to rabbit bone marrow stromal cells (BMSCs) by a tropism-modified human adenovirus serotype 5 (Ad5) vector as a novel therapeutic approach for spinal fusion. SUMMARY OF BACKGROUND DATA: Novel approaches are needed to improve the success of bone union after spinal fusion. One such approach is the ex vivo transfer of a gene encoding an osteoinductive factor to BMSCs which are subsequently reimplanted into the host. We have previously shown that heterologous expression of the Hoxc-8-interacting domain of Smad1 in the nuclei of osteoblast precursor cells is able to stimulate the expression of genes related to osteoblast differentiation and induce osteogenesis in vivo. Gene delivery vehicles based on human Ad5 are well suited for gene transfer for spinal fusion because they can mediate high-level, short-term gene expression. However, Ad5-based vectors with native tropism poorly transduce BMSCs, necessitating the use of vectors with modified tropism to achieve efficient gene transfer. METHODS: The gene encoding Smad1C was transferred to rabbit BMSCs by an Ad5 vector with native tropism or a vector retargeted to alphav integrins, which are abundantly expressed on rabbit BMSCs. Transduced BMSCs were maintained in osteoblastic differentiation medium for 30 days. Alkaline phosphatase activity was determined and cells stained for calcium deposition. As positive controls for osteogenesis, we used Ad5 vectors expressing bone morphogenetic protein 2. As negative controls, BMSCs were mock-transduced or transduced with an Ad5 vector expressing beta-galactosidase. In an immunocompetent rabbit model of spinal fusion, transduced BMSCs were coated onto absorbable gelatin sponge and implanted between decorticated transverse processes L6 and L7 of 8-week-old female New Zealand white rabbits. Animals were killed 4 weeks after implantation of the sponges, the fusion masses harvested and the area of new bone quantified using image analysis software. RESULTS: The Smad1C-expressing tropism-modified Ad5 vector mediated a significantly higher level of alkaline phosphatase activity and calcium deposition in transduced rabbit BMSCs than all other vectors. The rabbit BMSCs transduced ex vivo with the Smad1C-expressing tropism-modified Ad5 vector mediated a greater amount of new bone formation than BMSCs transduced with any other vector. CONCLUSIONS: Delivery of the Smad1C gene construct to BMSCs by an alphav integrin-targeted Ad5 vector shows promise for spinal fusion and other applications requiring the formation of new bone in vivo.


Asunto(s)
Adenoviridae/genética , Trasplante de Médula Ósea/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteína Smad1/genética , Fusión Vertebral/métodos , Células del Estroma/trasplante , Animales , Regeneración Ósea/genética , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Integrina alfaV/genética , Estructura Terciaria de Proteína/genética , Conejos , Células del Estroma/citología , Células del Estroma/metabolismo , Resultado del Tratamiento
8.
Mol Imaging ; 8(5): 264-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19796604

RESUMEN

We sought to develop a cancer-targeted, infectivity-enhanced oncolytic adenovirus that embodies a capsid-labeling fusion for noninvasive dual-modality imaging of ovarian cancer virotherapy. A functional fusion protein composed of fluorescent and nuclear imaging tags was genetically incorporated into the capsid of an infectivity-enhanced conditionally replicative adenovirus. Incorporation of herpes simplex virus thymidine kinase (HSV-tk) and monomeric red fluorescent protein 1 (mRFP1) into the viral capsid and its genomic stability were verified by molecular analyses. Replication and oncolysis were evaluated in ovarian cancer cells. Fusion functionality was confirmed by in vitro gamma camera and fluorescent microscopy imaging. Comparison of tk-mRFP virus to single-modality controls revealed similar replication efficiency and oncolytic potency. Molecular fusion did not abolish enzymatic activity of HSV-tk as the virus effectively phosphorylated thymidine both ex vivo and in vitro. In vitro fluorescence imaging demonstrated a strong correlation between the intensity of fluorescent signal and cytopathic effect in infected ovarian cancer cells, suggesting that fluorescence can be used to monitor viral replication. We have in vitro validated a new infectivity-enhanced oncolytic adenovirus with a dual-imaging modality-labeled capsid, optimized for ovarian cancer virotherapy. The new agent could provide incremental gains toward climbing the barriers for achieving conditionally replicated adenovirus efficacy in human trials.


Asunto(s)
Adenoviridae/metabolismo , Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/terapia , Adenoviridae/genética , Cápside/metabolismo , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/fisiología , Microscopía Fluorescente , Simplexvirus/enzimología , Timidina Quinasa/genética , Timidina Quinasa/fisiología , Proteína Fluorescente Roja
9.
J Med Virol ; 80(9): 1595-603, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18649343

RESUMEN

Malignant glioma, in particular glioblastoma multiforme (GBM), represents one of the most devastating cancers currently known and existing treatment regimens do little to change patient prognosis. Conditionally replicating adenoviral vectors (CRAds) represent attractive experimental anti-cancer agents with potential for clinical application. However, early protein products of the wild type adenovirus backbone--such as E1A--limit CRAds' replicative specificity. In this study, we evaluated the oncolytic potency and specificity of CRAds in which p300/CPB and/or pRb binding capacities of E1A were ablated to reduce non-specific replicative cytolysis. In vitro cytopathic assays, quantitative PCR analysis, Western blot, and flow cytometry studies demonstrate the superior anti-glioma efficacy of a double-mutated CRAd, Ad2/24CMV, which harbors mutations that reduce E1A binding to p300/CPB and pRb. When compared to its single-mutated and wild type counterparts, Ad2/24CMV demonstrated attenuated replication and cytotoxicity in representative normal human brain while displaying enhanced replicative cytotoxicity in malignant glioma. These results have implications for the development of double-mutated CRAd vectors for enhanced GBM therapy.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Glioma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Línea Celular , Humanos , Mutación , Replicación Viral
10.
Int J Oncol ; 31(5): 1177-85, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17912445

RESUMEN

Targeting gene expression to cancer cells remains a challenge for the development of gene and viral therapy for gliomas. Recent studies have highlighted transcriptional targeting as one of the possible solutions to overcome this limitation. In this context, melanoma associated antigens (MAAs) are usually over-expressed in brain tumors in comparison to normal brain tissue. For this reason, we investigated the use of the tyrosinase promoter as a transcriptional element to target oncolytic therapy for gliomas. Tyrosinase mRNA expression was evaluated by qRT-PCR in normal human brain tissue as well as in human glioma specimens. We found that this gene was significantly over-expressed in glioma cell lines and in primary glioma samples. Tyrosinase expression correlated with the grade of the tumor (p-value range: 0.05-0.001). Furthermore, transfection of several cell cultures with human and mouse tyrosinase promoters driving a luciferase reporter gene confirmed the activity of this promoter in mouse and human cells. To evaluate whether tyrosinase-activated conditionally replicative adenoviruses (CRAds) could induce toxicity in glioma cells, two vectors (Ad h/m and Ad24TYR) were tested in a mouse glioma model. C57BL/6 mice underwent intracranial injection of tumor cell line GL261. Survival was used to evaluate efficacy of the tested vectors. Mice receiving 1 x 10(9) MOI of Ad h/m and Ad24TYR following intracranial tumor implants had a median survival of 46+/-3 days (p<0.05); in contrast, those treated with medium had a median survival of 31+/-2 days. These results suggest that injection of tyrosinase CRAds leads to prolongation of survival in mice with experimental brain tumors. The tyrosinase promoter stands as a proof of principle of the potential use of MAA over-expression patterns for targeting novel anti-glioma therapies.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Vectores Genéticos/genética , Glioma/terapia , Monofenol Monooxigenasa/genética , Regiones Promotoras Genéticas , Animales , Glioma/enzimología , Humanos , Ratones , Ratones Endogámicos C57BL , Replicación Viral
11.
Mol Biotechnol ; 36(1): 71-80, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17827541

RESUMEN

Vectors based on human adenovirus serotypes 2 (Ad2) and 5 (Ad5) of species C possess a number of features that have favored their widespread employment for gene delivery both in vitro and in vivo. However, the use of recombinant Ad2- and Ad5-based vectors for gene therapy also suffers from a number of disadvantages. These vectors possess the tropism of the parental viruses, which infect all cells that possess the appropriate surface receptors, precluding the targeting of specific cell types. Conversely, some cell types that represent important targets for gene transfer express only low levels of the cellular receptors, which lead to inefficient infection. Another major disadvantage of Ad2- and Ad5-based vectors in vivo is the elicitation of both an innate and an acquired immune response. Considerable attention has therefore been focused on strategies to overcome these limitations, thereby permitting the full potential of adenoviral vectors to be realized.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Vectores Genéticos , Adenoviridae/química , Infecciones por Adenoviridae/metabolismo , Animales , Humanos
12.
Cancer Biol Ther ; 5(12): 1647-53, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17106248

RESUMEN

Clinical studies of replicating adenoviruses for the treatment of cancer have demonstrated their safety but have yielded disappointing results, indicating the need for new strategies to improve their efficacy. We hypothesized that the efficacy of a replicating adenovirus could be improved by expression of tissue inhibitor of metalloproteinases-2 (TIMP-2), a 21-kDa unglycosylated secretory protein. TIMP-2 specifically inhibits the active forms of a number of matrix metalloproteinases (MMPs) that play a role in the degradation of basement membranes and the extracellular matrix and are therefore involved in the control of the growth, invasion and metastasis of tumor cells, as well as angiogenesis. In addition, TIMP-2 can abrogate tumor growth and angiogenesis by a variety of mechanisms independent of MMP inhibition. In this study, we demonstrate that expression of TIMP-2 enhanced the antitumor efficacy of a replicating adenovirus in vivo, by reducing both tumor growth and angiogenesis.


Asunto(s)
Adenovirus Humanos/fisiología , Neoplasias de la Mama/virología , Neoplasias/prevención & control , Inhibidor Tisular de Metaloproteinasa-2/genética , Replicación Viral/fisiología , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/genética , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citomegalovirus/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/prevención & control , Regiones Promotoras Genéticas , Trasplante Heterólogo
13.
Virus Res ; 116(1-2): 185-95, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16293334

RESUMEN

The utility of adenovirus serotype 5 (Ad5)-based vectors for gene therapy applications would be improved by cell-specific targeting. However, strategies to redirect Ad5 vectors to alternate cellular receptors via replacement of the capsid fiber protein have often resulted in structurally unstable vectors. In view of this, we hypothesized that the selection of modified adenoviruses during their rescue and propagation would be a straightforward approach that guarantees the generation of functional, targeted vectors. Based on our first generation fiber-fibritin molecule, several new chimeric fibers containing variable amounts of fibritin and the Ad5 fiber shaft were analyzed via a new scheme for Ad vector selection. Our selected chimera, composed of the entire Ad5 fiber shaft fused to the 12th coiled-coil segment of fibritin, is capable of efficient capsid incorporation and ligand display. Moreover, transduction by the resultant vector is independent of the expression of the native Ad5 receptor. The incorporation of the Fc-binding domain of Staphylococcus aureus protein A at the carboxy terminus of this chimeric fiber facilitates targeting of the vector to a variety of cellular receptors by means of coupling with monoclonal antibodies. In addition, we have concluded that Ad5 vectors incorporating individual targeting ligands require individual optimization of the fiber-fibritin chimera, which may be accomplished by selecting the optimal fiber-fibritin variant at the stage of rescue of the virus in cells of interest, as described herein.


Asunto(s)
Adenoviridae/genética , Anticuerpos Monoclonales/metabolismo , Vectores Genéticos/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/fisiología , Línea Celular , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Ligandos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo , Proteínas Virales/genética , Proteínas Virales/fisiología
14.
J Bone Miner Res ; 20(10): 1858-66, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16160744

RESUMEN

UNLABELLED: Spaceflight, aging, and disuse lead to reduced BMD. This study shows that overexpression of constitutively active RhoA restores actin cytoskeletal arrangement, enhances the osteoblastic phenotype, and suppresses the adipocytic phenotype of human mesenchymal stem cells cultured in modeled microgravity. INTRODUCTION: Reduced BMD during spaceflight is partly caused by reduced bone formation. However, mechanisms responsible for this bone loss remain unclear. We have previously shown reduced osteoblastogenesis and enhanced adipogenesis of human mesenchymal stem cells (hMSCs) cultured in modeled microgravity (MMG). The small GTPase, RhoA, regulates actin stress fiber formation and has been implicated in the lineage commitment of hMSCs. We examined the effects of MMG on actin cytoskeletal organization and RhoA activity and the ability of constitutively active RhoA to reverse these effects. MATERIALS AND METHODS: hMSCs were seeded onto plastic microcarrier beads at a density of 10(6) and allowed to form aggregates in DMEM containing 10% FBS for 7 days. Aggregates were incubated in DMEM containing 2% FBS for 6 h with or without an adenoviral vector containing constitutively active RhoA at a multiplicity of infection (moi) of 500 and allowed to recover in 10% FBS for 24 h. Cells were transferred to the rotary cell culture system to model microgravity or to be maintained at normal gravity for 7 days in DMEM, 10% FBS, 10 nM dexamethasone, 10 mM beta-glycerol phosphate, and 50 muM ascorbic acid 2-phosphate. RESULTS: F-actin stress fibers are disrupted in hMSCs within 3 h of initiation of MMG and are completely absent by 7 days, whereas monomeric G-actin is increased. Because of the association of G-actin with lipid droplets in fat cells, the observed 310% increase in intracellular lipid accumulation in hMSCs cultured in MMG was not unexpected. Consistent with these changes in cellular morphology, 7 days of MMG significantly reduces RhoA activity and subsequent phosphorylation of cofilin by 88+/-2% and 77+/-9%, respectively. Importantly, introduction of an adenoviral construct expressing constitutively active RhoA reverses the elimination of stress fibers, significantly increases osteoblastic gene expression of type I collagen, alkaline phosphatase, and runt-related transcription factor 2, and suppresses adipocytic gene expression of leptin and glucose transporter 4 in hMSCs cultured in MMG. CONCLUSION: Suppression of RhoA activity during MMG represents a novel mechanism for reduced osteoblastogenesis and enhanced adipogenesis of hMSCs.


Asunto(s)
Adipogénesis , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Fibras de Estrés/metabolismo , Ingravidez/efectos adversos , Proteína de Unión al GTP rhoA/metabolismo , Envejecimiento/metabolismo , Densidad Ósea , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteoblastos/metabolismo , Vuelo Espacial
15.
Biochim Biophys Acta ; 1575(1-3): 1-14, 2002 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-12020813

RESUMEN

Replication-defective vectors based on human adenovirus serotypes 2 and 5 (Ad2 and Ad5) possess a number of attributes which favor their use as gene delivery vehicles in gene therapy applications. However, the widespread distribution of the primary cellular receptor for Ad, the coxsackievirus and adenovirus receptor (CAR), allows Ad vectors to infect a broad range of cells in the host. Conversely, a number of tissues which represent important targets for gene therapy, such as the airway epithelium and cancer cells, are refractory to Ad infection due a paucity of CAR. Thus, there is a strong rationale for the development of CAR-independent Ad vectors capable of enhanced specificity and efficiency of gene transfer to target cells. In this article we review the approaches which have been employed to generate tropism-modified Ad vectors. These targeting strategies have led to improvements in the safety and efficacy of Ad vectors and have the potential to yield an increased therapeutic benefit in the human clinical context.


Asunto(s)
Adenoviridae , Marcación de Gen , Técnicas de Transferencia de Gen , Vectores Genéticos , Infecciones por Adenoviridae , Animales , Terapia Genética , Humanos
16.
Cancer Gene Ther ; 12(5): 475-86, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15706356

RESUMEN

The use of adenovirus type 5 (Ad5) for cancer therapy is limited by deficiency of its primary cell attachment receptor, coxsackie and adenovirus receptor (CAR), on cancer cells. Ad5 retargeting to alternate receptors through fiber genetic modification can be used to circumvent CAR dependence of its tropism, and thereby achieve infectivity enhancement. Here we propose and test a novel "complex mosaicism" approach for fiber modification, which combines serotype chimerism with peptide ligand(s) incorporation in a single-fiber molecule. We incorporated integrin-binding peptide RGD-4C in the HI-loop, at the carboxy (C)-terminus, or both locales of the Ad3 knob, in the context of Ad5/3 chimera fiber in order to retarget simultaneously the Ad vector to integrins and Ad3 receptors. The infectivity enhancement of the fiber modifications was assessed in various cancer cell lines as cancer-targeting models. Replication-defective complex mosaic Ad-luc vectors bearing chimeric fiber (F.5/3), with or without C-terminal RGD-modification of Ad3 knob, demonstrated up to 55-fold gene transfer increase in bladder cancer cell lines. Although this augmentation was primarily due to Ad3 receptor targeting, some contribution of RGD-mediated integrin-targeting was also observed, suggesting that complex mosaic modification can function in a dual-receptor targeting via a single Ad3 fiber knob.


Asunto(s)
Adenoviridae/genética , Proteínas de la Cápside/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Humanos , Integrinas/metabolismo , Oligopéptidos/genética , Receptores Virales/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
17.
Cancer Biol Ther ; 4(3): 289-94, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15753654

RESUMEN

Dendritic cells (DCs) are a central element in the development of antigen-specific immune responses. The lack of a specific and efficient technique for the in vivo delivery of antigens to DCs remains a major obstacle limiting a vaccine's ability to induce an effective immune response. The efficacy of adenoviral (Ad) vectors in this regard can be enhanced through alterations in vector tropism such that DC-targeted transduction is achieved. Here, the efficiency of DC transduction by Ad vectors retargeted to DC-specific ICAM-3 grabbing nonintegrin (DC-SIGN) was studied and compared to that of Ad vectors retargeted through CD40. A comparable and significant enhancement of gene transfer to monocyte derived DCs (MDDCs) was accomplished by means of an Ad vector harboring the Fc-binding domain of Staphylococcus aureus protein A in combination with antibodies to DC-SIGN or to CD40 or with fused complexes of human Ig-Fc with their natural ligands, i.e., ICAM-3 or CD40L, respectively. Whereas CD40-targeted Ad transduction resulted in a more profound phenotypic DC maturation, DC-SIGN- and CD40-targeted Ad both induced similar levels of IL-12 secretion. These data demonstrate the usefulness of DC-SIGN as a DC-restricted targeting motif for Ad-mediated vaccination strategies.


Asunto(s)
Adenoviridae/genética , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/inmunología , Vectores Genéticos/genética , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción Genética/métodos , Antígenos CD40/genética , Vacunas contra el Cáncer/genética , Línea Celular , Humanos , Inmunoterapia Adoptiva , Monocitos/inmunología
18.
Clin Cancer Res ; 8(6): 1915-23, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12060636

RESUMEN

PURPOSE: Several barriers that collectively restrict gene delivery by viral vectors in vivo have been described. Previously, we identified soluble chondroitin sulfate-proteoglycans/glycosaminoglycans in malignant pleural effusions (MPEs) as inhibitors of retroviral vector transduction. Soluble components of MPE also inhibited adenoviral (Ad) gene transfer, and the factors were characteristically filterable, titrable, stable at 56 degrees C, and blocked the binding of Ad to target cells. Depleting immunoglobulin from MPE, partially reversed the block to Ad transduction, instigating a search for additional factors that bound Ad in MPE. EXPERIMENTAL DESIGN: Vector-protein interactions were identified after the resolution of MPE-components by SDS-PAGE. Viral overlays and immunoblots delineated significant interactions, and the potential relevance of those interactions was tested in transduction efficiency bioassays. RESULTS: Immunoglobulin is the predominant factor inhibiting Ad gene transfer in MPE. Albumin also interacted with Ad, although at predicted serum concentrations, it did not effect Ad transduction efficiency in vitro. Soluble coxsackievirus-Ad receptor (sCAR) was then identified in MPE. In a survey of 18 MPE, the mean concentration of sCAR was variable and estimated to be 3.51 +/- 5.02 ng/ml by ELISA. The impact of sCAR on transduction efficiency in this milieu was next assessed. Whereas immunodepletion of sCAR from MPE by affinity chromatography resulted in enhanced gene transfer within MPE, the inhibition of adenoviral gene transfer was not evident when the predicted concentrations of recombinant sCAR were added into the transduction medium. CONCLUSIONS: These studies indicate that, in addition to anti-Ad antibodies, other specific and nonspecific factors interact with viral vectors and may impair gene transfer in the tumor milieu. The presence of sCAR in MPE puts forward the notion that in certain contexts (e.g., within the extracellular matrix of solid tumors) the concentrations of secreted (or shed) CAR may be high enough to effectively compete with Ad gene delivery.


Asunto(s)
Adenoviridae/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Enterovirus/genética , Técnicas de Transferencia de Gen , Neoplasias Pulmonares/metabolismo , Receptores Virales/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Terapia Genética , Vectores Genéticos , Humanos , Operón Lac/fisiología , Neoplasias Pulmonares/genética , Ratones , Derrame Pleural/metabolismo , Receptores Virales/inmunología , Células Tumorales Cultivadas
19.
Hum Gene Ther ; 15(5): 509-18, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15144580

RESUMEN

Approaches to alter the native tropism of adenoviruses (Ads) are beneficial to increase their efficacy and safety profile. Liver tropism is important with regard to potential clinical toxicity in humans. Ad5/3 chimeras in which the Ad5 knob is substituted by the Ad3 knob, such as Ad5/3luc1, have been recently shown to increase infectivity of ovarian cancer cell lines and primary tumor cells, which express low levels of the coxsackie-adenovirus receptor (CAR), without increasing infectivity of liver cells. A novel strategy to address the problem of liver uptake and improve the tumor/liver ratio is genetic replacement of the Ad fiber shaft. Ad5.Ad3.SH.luc1 is an Ad5-based vector that contains the fiber shaft from Ad serotype 3 but the fiber knob from Ad serotype 5. To compare tumor/liver of Ad5.Ad3.SH.luc1 and Ad5/3luc1 in vivo, we created three different tumor and treatment models of ovarian cancer in mice, simulating intraperitoneal and intravenous administration of tumors. Ad5.Ad3.SH.luc1 displayed the lowest liver tropism of all viruses in all models tested. Intravenous administration of all viruses resulted in higher tumor transduction rates compared to intraperitoneal administration. Genetic shortening of the Ad5 fiber shaft significantly increases relative tumor/liver gene transfer. This could improve the effective tumor dose and reduce side effects, thereby increasing the bioavailability of therapeutic agents.


Asunto(s)
Adenovirus Humanos/genética , Técnicas de Transferencia de Gen , Hígado/metabolismo , Neoplasias Ováricas/terapia , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Femenino , Terapia Genética , Vectores Genéticos , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Subcutáneas , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Especificidad de Órganos , Proteínas Recombinantes/metabolismo , Recombinación Genética , Serotipificación , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Gene Ther ; 10(2): 121-4, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12536200

RESUMEN

As an initial assessment of the feasibility of employing the adenovirus serotype 3 (Ad3) fiber knob as a locale for introducing a tropism-modifying motif, we generated an adenoviral vector containing a six-histidine tag genetically fused to the carboxy-terminus of the Ad3 fiber knob. The heterologous tag proved to be accessible for binding in the context of the virion and, moreover, had rendered the modified vector capable of mediating gene transfer through an artificial, non-Ad3 receptor.


Asunto(s)
Adenoviridae/genética , Cápside/fisiología , Vectores Genéticos/genética , Adenoviridae/metabolismo , Anticuerpos Monoclonales/metabolismo , Secuencia de Bases , Citomegalovirus/genética , ADN Viral/análisis , ADN Viral/genética , Ensayo de Inmunoadsorción Enzimática , Ingeniería Genética/métodos , Proteínas Fluorescentes Verdes , Histidina/genética , Ligandos , Luciferasas/genética , Proteínas Luminiscentes/genética , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA