Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Gene Ther ; 24(3): 176-186, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27996967

RESUMO

We evaluated late effects of AdhAQP1 administration in five subjects in a clinical trial for radiation-induced salivary hypofunction (http://www.clinicaltrials.gov/ct/show/NCT00372320?order=). All were identified as initially responding to human aquaporin-1 (hAQP1) gene transfer. They were followed for 3-4 years after AdhAQP1 delivery to one parotid gland. At intervals we examined salivary flow, xerostomic symptoms, saliva composition, vector presence and efficacy in the targeted gland, clinical laboratory data and adverse events. All displayed marked increases (71-500% above baseline) in parotid flow 3-4.7 years after treatment, with improved symptoms for ~2-3 years. There were some changes in [Na+] and [Cl-] consistent with elevated salivary flow, but no uniform changes in secretion of key parotid proteins. There were no clinically significant adverse events, nor consistent negative changes in laboratory parameters. One subject underwent a core needle biopsy of the targeted parotid gland 3.1 years post treatment and displayed evidence of hAQP1 protein in acinar, but not duct, cell membranes. All subjects responding to hAQP1 gene transfer initially had benefits for much longer times. First-generation adenoviral vectors typically yield transit effects, but these data show beneficial effects can continue years after parotid gland delivery.


Assuntos
Aquaporina 1/genética , Terapia Genética/efeitos adversos , Xerostomia/terapia , Adenoviridae/genética , Aquaporina 1/metabolismo , Cloretos/metabolismo , Vetores Genéticos/genética , Humanos , Pessoa de Meia-Idade , Radioterapia/efeitos adversos , Glândulas Salivares/metabolismo , Sódio/metabolismo , Xerostomia/etiologia
2.
Oral Dis ; 23(6): 801-808, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28326646

RESUMO

OBJECTIVE: The goals of this study were to (i) establish a useful miniature pig (minipig) model for irradiation-induced oral mucositis and (ii) evaluate the effect of Tempol to prevent its development. METHODS AND MATERIALS: Minipigs were irradiated with 6 Gy for five consecutive days targeting the entire oral cavity. To prevent radiation damage, minipigs were treated with 30 mg kg-1 Tempol 10 min before irradiation (n = 4), while the radiation-alone group was similarly injected with saline (n = 4). Lesions were graded using an oral mucositis score and visual inspection every 3 days, and biopsy of multiple sites was performed at day 18. Weight and chest and abdominal circumferences were measured every 3 days. RESULTS: Lesions began about 12 days after the first irradiation fraction and healed about 30 days after irradiation. Epithelial thickness was calculated on the lingual and buccal mucosa on the 18th day after the first irradiation fraction. Tempol provided modest protection from ulceration after irradiation using this treatment strategy. CONCLUSIONS: This study established a useful large animal model for irradiation-induced oral mucositis and showed modest beneficial effects of Tempol in limiting tissue damage. The latter finding may be potentially valuable in preventing oral mucositis in patients receiving irradiation for head and neck cancers.


Assuntos
Óxidos N-Cíclicos/uso terapêutico , Lesões por Radiação/prevenção & controle , Protetores contra Radiação/uso terapêutico , Estomatite/prevenção & controle , Animais , Modelos Animais de Doenças , Masculino , Radioterapia/efeitos adversos , Marcadores de Spin , Suínos
3.
Oral Dis ; 23(3): 337-346, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27886428

RESUMO

OBJECTIVES: The purpose of this study was to examine the humoral and cellular immune reactivity to adenoviral vector (AdhAQP1) administration in the human parotid gland over the first 42 days of a clinical gene therapy trial. METHODS: Of eleven treated subjects, five were considered as positive responders (Baum et al, 2012). Herein, we measured serum neutralizing antibody titers, circulating cytotoxic lymphocytes, and lymphocyte proliferation in peripheral blood mononuclear cells. Additionally, after adenoviral vector stimulation of lymphocyte proliferation, we quantified secreted cytokine levels. RESULTS: Responders showed little to modest immune reactivity during the first 42 days following gene transfer. Additionally, baseline serum neutralizing antibody titers to serotype 5-adenovirus generally were not predictive of a subject's response to parotid gland administration of AdhAQP1. Cytokine profiling from activated peripheral blood mononuclear cells could not distinguish responders and non-responders. CONCLUSIONS: The data are the first to describe immune responses after adenoviral vector administration in a human parotid gland. Importantly, we found that modest (2-3 fold) changes in systemic cell-mediated immune reactivity did not preclude positive subject responses to gene transfer. However, changes beyond that level likely impeded the efficacy of gene transfer.


Assuntos
Adenoviridae/imunologia , Anticorpos Neutralizantes/sangue , Vetores Genéticos/imunologia , Linfócitos T Citotóxicos , Idoso , Aquaporina 1/genética , Proliferação de Células , Citocinas/sangue , DNA Complementar/genética , Feminino , Terapia Genética , Humanos , Imunidade Celular , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Glândula Parótida/virologia , Linfócitos T Citotóxicos/fisiologia
4.
Gene Ther ; 23(7): 572-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26966862

RESUMO

Head and neck irradiation (IR) during cancer treatment causes by-stander effects on the salivary glands leading to irreversible loss of saliva secretion. The mechanism underlying loss of fluid secretion is not understood and no adequate therapy is currently available. Delivery of an adenoviral vector encoding human aquaporin-1 (hAQP1) into the salivary glands of human subjects and animal models with radiation-induced salivary hypofunction leads to significant recovery of saliva secretion and symptomatic relief in subjects. To elucidate the mechanism underlying loss of salivary secretion and the basis for AdhAQP1-dependent recovery of salivary gland function we assessed submandibular gland function in control mice and mice 2 and 8 months after treatment with a single 15-Gy dose of IR (delivered to the salivary gland region). Salivary secretion and neurotransmitter-stimulated changes in acinar cell volume, an in vitro read-out for fluid secretion, were monitored. Consistent with the sustained 60% loss of fluid secretion following IR, a carbachol (CCh)-induced decrease in acinar cell volume from the glands of mice post IR was transient and attenuated as compared with that in cells from non-IR age-matched mice. The hAQP1 expression in non-IR mice induced no significant effect on salivary fluid secretion or CCh-stimulated cell volume changes, except in acinar cells from 8-month group where the initial rate of cell shrinkage was increased. Importantly, the expression of hAQP1 in the glands of mice post IR induced recovery of salivary fluid secretion and a volume decrease in acinar cells to levels similar to those in cells from non-IR mice. The initial rates of CCh-stimulated cell volume reduction in acinar cells from hAQP1-expressing glands post IR were similar to those from control cells. Altogether, the data suggest that expression of hAQP1 increases the water permeability of acinar cells, which underlies the recovery of fluid secretion in the salivary glands functionally compromised post IR.


Assuntos
Células Acinares/metabolismo , Aquaporina 1/genética , Tamanho Celular , Glândulas Salivares/metabolismo , Células Acinares/citologia , Células Acinares/efeitos da radiação , Adenoviridae/genética , Animais , Aquaporina 1/metabolismo , Linhagem Celular , Células Cultivadas , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Camundongos , Radiação Ionizante , Ratos , Saliva/metabolismo , Glândulas Salivares/citologia , Glândulas Salivares/efeitos da radiação
5.
Gene Ther ; 22(9): 758-66, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26177970

RESUMO

In 2012, we reported that 5 out of 11 subjects in a clinical trial (NCT00372320) administering AdhAQP1 to radiation-damaged parotid glands showed increased saliva flow rates and decreased symptoms over the initial 42 days. AdhAQP1 is a first-generation, E1-deleted, replication-defective, serotype 5 adenoviral vector encoding human aquaporin-1 (hAQP1). This vector uses the human cytomegalovirus enhancer/promoter (hCMVp). As subject peak responses were at times much longer (7-42 days) than expected, we hypothesized that the hCMVp may not be methylated in human salivary gland cells to the extent previously observed in rodent salivary gland cells. This hypothesis was supported in human salivary gland primary cultures and human salivary gland cell lines after transduction with AdhAQP1. Importantly, hAQP1 maintained its function in those cells. Conversely, when we transduced mouse and rat cell lines in vitro and submandibular glands in vivo with AdhAQP1, the hCMVp was gradually methylated over time and associated with decreased hAQP1 expression and function in vitro and decreased hAQP1 expression in vivo. These data suggest that the hCMVp in AdhAQP1was probably not methylated in transduced human salivary gland cells of responding subjects, resulting in an unexpectedly longer functional expression of hAQP1.


Assuntos
Aquaporina 1/metabolismo , Citomegalovirus/genética , Expressão Gênica , Regiões Promotoras Genéticas , Glândulas Salivares/metabolismo , Transdução Genética , Animais , Linhagem Celular , Humanos , Metilação , Camundongos , Ratos
6.
Gene Ther ; 21(10): 866-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25030610

RESUMO

Patients frequently experience a loss of salivary function following irradiation (IR) for the treatment of an oral cavity and oropharyngeal cancer. Herein, we tested if transfer of fibroblast growth factor-2 (FGF2) cDNA could limit salivary dysfunction after fractionated IR (7.5 or 9 Gy for 5 consecutive days to one parotid gland) in the miniature pig (minipig). Parotid salivary flow rates steadily decreased by 16 weeks post-IR, whereas blood flow in the targeted parotid gland began to decrease ~3 days after beginning IR. By 2 weeks, post-IR salivary blood flow was reduced by 50%, at which point it remained stable for the remainder of the study. The single preadministration of a hybrid serotype 5 adenoviral vector encoding FGF2 (AdLTR2EF1a-FGF2) resulted in the protection of parotid microvascular endothelial cells from IR damage and significantly limited the decline of parotid salivary flow. Our results suggest that a local treatment directed at protecting salivary gland endothelial cells may be beneficial for patients undergoing IR for oral cavity and oropharyngeal cancer.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Vetores Genéticos/administração & dosagem , Glândula Parótida/fisiopatologia , Lesões Experimentais por Radiação/prevenção & controle , Animais , Dependovirus/genética , Células Endoteliais/metabolismo , Células Endoteliais/efeitos da radiação , Fator 2 de Crescimento de Fibroblastos/genética , Terapia Genética , Glândula Parótida/citologia , Glândula Parótida/efeitos da radiação , Lesões Experimentais por Radiação/patologia , Saliva/citologia , Saliva/efeitos da radiação , Suínos , Porco Miniatura
7.
Gene Ther ; 17(1): 50-60, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19759566

RESUMO

Salivary glands are potentially useful target sites for multiple clinical applications of gene transfer. Previously, we have shown that serotype 2 adeno-associated viral (AAV2) vectors lead to stable gene transfer in the parotid glands of rhesus macaques. As AAV5 vectors result in considerably greater transgene expression in murine salivary glands than do AAV2 vectors, herein we have examined the use of AAV5 vectors in macaques at two different doses (n = 3 per group; 10(10) or 3 x 10(11) particles per gland). AAV5 vector delivery, as with AAV2 vectors, led to no untoward clinical, hematological or serum chemistry responses in macaques. The extent of AAV5-mediated expression of rhesus erythropoietin (RhEpo) was dose-dependent and similar to that seen with an AAV2 vector. However, unlike results with the AAV2 vector, AAV5 vector-mediated RhEpo expression was transient. Maximal expression peaked at day 56, was reduced by approximately 80% on day 84 and thereafter remained near background levels until day 182 (end of experiment). Quantitative PCR studies of high-dose vector biodistribution at this last time point showed much lower AAV5 copy numbers in the targeted parotid gland (approximately 1.7%) than found with the same AAV2 vector dose. Molecular analysis of the conformation of vector DNA indicated a markedly lower level of concatamerization for the AAV5 vector compared with that of a similar AAV2 vector. In addition, cellular immunological studies suggest that host response differences may occur with AAV2 and AAV5 vector delivery at this mucosal site. The aggregate data indicate that results with AAV5 vectors in murine salivary glands apparently do not extend to macaque glands.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Glândula Parótida/metabolismo , Animais , Terapia Genética/métodos , Macaca mulatta , Glândula Parótida/virologia , Transdução Genética , Transgenes
8.
Oral Dis ; 16(3): 269-77, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20374510

RESUMO

OBJECTIVES: Salivary glands are useful target organs for local and systemic gene therapeutics. For such applications, the regulation of transgene expression is important. Previous studies by us in murine submandibular glands showed that a rapamycin transcriptional regulation system in a single serotype 2, adeno-associated viral (AAV2) vector was effective for this purpose. This study evaluated if such a vector was similarly useful in rhesus macaque parotid glands. METHODS: A recombinant AAV2 vector (AAV-TF-RhEpo-2.3w), encoding rhesus erythropoietin (RhEpo) and a rapamycin-inducible promoter, was constructed. The vector was administered to macaques at either of two doses [1.5 x 10(11) (low dose) or 1.5 x 10(12) (high dose) vector genomes] via cannulation of Stensen's duct. Animals were followed up for 12-14 weeks and treated at intervals with rapamycin (0.1 or 0.5 mg kg(-1)) to induce gene expression. Serum chemistry, hematology, and RhEpo levels were measured at interval. RESULTS: AAV-TF-RhEpo-2.3w administration led to low levels of rapamycin-inducible RhEpo expression in the serum of most macaques. In five animals, no significant changes were seen in serum chemistry and hematology values over the study. One macaque, however, developed pneumonia, became anemic and subsequently required euthanasia. After the onset of anemia, a single administration of rapamycin led to significant RhEpo production in this animal. CONCLUSION: Administration of AAV-TF-RhEpo-2.3w to macaque parotid glands was generally safe, but led only to low levels of serum RhEpo in healthy animals following rapamycin treatment.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/administração & dosagem , Glândula Parótida/metabolismo , Sirolimo/farmacologia , Transdução Genética , Adenoviridae/genética , Animais , Relação Dose-Resposta a Droga , Eritropoetina/sangue , Eritropoetina/genética , Eritropoetina/metabolismo , Macaca mulatta , Masculino , Regiões Promotoras Genéticas , Proteínas Recombinantes , Transgenes
9.
Oral Dis ; 15(1): 76-81, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18992019

RESUMO

OBJECTIVES: Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a multifunctional cytokine that has pro-apoptotic, pro-angiogenic and pro-inflammatory effects. In liver, TWEAK leads to proliferation of progenitor oval cells, but not of mature hepatocytes. This study evaluated the hypothesis that TWEAK overexpression in salivary glands would lead to the proliferation of a salivary progenitor cell. METHODS: A recombinant, serotype 5 adenoviral vector encoding human TWEAK, AdhTWEAK, was constructed, initially tested in vitro, and then administered to male Balb/c mice via cannulation of Wharton's duct. TWEAK expression in vivo was monitored as protein secreted into saliva and serum by enzyme-linked immunosorbent assays. Salivary cell proliferation was monitored by proliferating cell nuclear antigen staining and apoptosis was monitored using TUNEL staining. RESULTS: AdhTWEAK administration led to a dose-dependent, transient TWEAK protein expression, detected primarily in saliva. Salivary epithelial cell proliferation was generalized, peaking on approximately days 2 and 3. TWEAK expression had no detectable effect on apoptosis of salivary epithelial cells. CONCLUSION: Transient overexpression of TWEAK in murine salivary glands leads to a general proliferation of epithelial cells vs a selective stimulation of a salivary progenitor cell.


Assuntos
Apoptose/genética , Células Epiteliais/patologia , Regulação da Expressão Gênica/genética , Glândula Submandibular/patologia , Fatores de Necrose Tumoral/genética , Animais , Proliferação de Células , Citocina TWEAK , Vetores Genéticos/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Antígeno Nuclear de Célula em Proliferação/análise , Saliva/química , Ductos Salivares/patologia , Células-Tronco/patologia , Fatores de Tempo , Fatores de Necrose Tumoral/análise , Fatores de Necrose Tumoral/sangue
10.
Gene Ther ; 13(7): 594-601, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16341060

RESUMO

We previously demonstrated that recombinant adeno-associated virus vectors based on serotype 2 (rAAV2) can direct transgene expression in salivary gland cells in vitro and in vivo. However, it is not known how other rAAV serotypes perform when infused into salivary glands. The capsids of serotypes 4 and 5 are distinct from rAAV2 and from each other, suggesting that they may direct binding and entry into different cell types. In the present study, we investigated the tropisms, transduction efficiencies, and antibody response to AAV vectors based on AAV serotypes 2, 4, and 5. Administration of rAAV2beta-galactosidase (betagal), rAAV4betagal, or rAAV5betagal to murine submandibular salivary glands by retrograde ductal instillation resulted in efficient transduction of salivary epithelial cells, with AAV4 and AAV5 producing 2.3 and 7.3 times more betagal activity compared with AAV2. Improved transduction with AAV5 was confirmed by QPCR of DNA extracted from glands and immunohistochemical staining for transgene expression. Like AAV2, AAV5 primarily transduced striated and intercalated ductal cells. AAV4 transduction was evident in striated, intercalated, and excretory ductal cells, as well as in convoluted granular tubules. In keeping with the encapsulated nature of the salivary gland, the majority of persistent viral genomes were found in the gland and not in other tissues. Neutralizing antibodies (NABs) found in the serum of virus-infused animals were serotype specific and there was no crossreactivity between serotypes. No NABs were detected in saliva but sialic acid conjugates present in saliva could neutralize AAV4 at low dilutions. Together our data suggest that because of differences in receptor binding and transduction pathways, other serotypes may have improved utility as gene transfer vectors in the salivary gland and these differences could be exploited in gene therapy applications.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glândula Submandibular/metabolismo , Transdução Genética/métodos , Animais , Anticorpos Antivirais/sangue , Dependovirus/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Expressão Gênica , Vetores Genéticos/genética , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Parvoviridae/imunologia , Infecções por Parvoviridae/virologia , Saliva/imunologia , Sorotipagem , Glândula Submandibular/virologia , Transgenes , beta-Galactosidase/análise , beta-Galactosidase/genética
11.
Ann Rheum Dis ; 65(2): 195-200, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15975969

RESUMO

BACKGROUND: Sjögren's syndrome (SS), an autoimmune exocrinopathy mainly affecting lachrymal and salivary glands, results in ocular and oral dryness (keratoconjunctivitis sicca and xerostomia). The aetiology and pathogenesis are largely unknown; currently, only palliative treatment is available. OBJECTIVE: To determine whether gene transfer of vasoactive intestinal peptide (VIP), based on its immunomodulatory properties, might be useful in the management of SS. METHODS: A recombinant serotype 2 adeno-associated virus encoding the human VIP transgene (rAAV2hVIP) was constructed and its efficacy tested in the female non-obese diabetic (NOD) mouse model for SS after retrograde instillation in submandibular glands (SMGs). 10(10) particles/gland of rAAV2hVIP or rAAV2LacZ (encoding beta-galactosidase; control vector) were administered at 8 weeks of age (before sialadenitis onset). Salivary flow rates were determined before vector delivery and at time of death (16 weeks). After death, saliva, serum, and SMGs were harvested. Salivary output, inflammatory infiltrates (focus scores), VIP protein expression, cytokine profile, and serum anti-VIP antibodies were analysed. RESULTS: rAAV2hVIP significantly improved the salivary flow, increased SMG and serum expression of VIP, and reduced SMG cytokines interleukin (IL) 2, IL10, IL12 (p70), and tumour necrosis factor alpha, and serum RANTES, compared with the control vector. No difference in focus scores or apoptotic rates was found; neutralising antibodies were not detected. CONCLUSIONS: Local delivery of rAAV2hVIP can have disease modifying and immunosuppressive effects in SMGs of the NOD mouse model of SS. The new strategy of employing VIP prophylactically may be useful for both understanding and managing the salivary component of SS.


Assuntos
Terapia Genética/métodos , Síndrome de Sjogren/terapia , Transdução Genética/métodos , Peptídeo Intestinal Vasoativo/genética , Adenoviridae/genética , Animais , Anticorpos/sangue , Apoptose , Citocinas/análise , Citocinas/sangue , Células Epiteliais/patologia , Feminino , Expressão Gênica , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Óperon Lac , Camundongos , Camundongos Endogâmicos NOD , Modelos Animais , Síndrome de Sjogren/imunologia , Síndrome de Sjogren/patologia , Glândula Submandibular/imunologia , Glândula Submandibular/patologia , Transgenes , Peptídeo Intestinal Vasoativo/análise , Peptídeo Intestinal Vasoativo/sangue
12.
Oral Dis ; 12(6): 566-72, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17054769

RESUMO

OBJECTIVES: The non-obese diabetic (NOD) mouse is not only a widely used model for diabetes mellitus type I, but also for the chronic autoimmune disease Sjögren's syndrome (SS), mainly affecting salivary and lacrimal glands. We studied the efficacy of local recombinant serotype 2 adeno-associated viral (rAAV2) vector transfer of immunomodulatory transgenes to alter the SS-like disease in NOD mice. Data collected over a 2-year period indicated a changing SS phenotype in these mice and this phenomenon was investigated. METHODS: 10(10) particles rAAV2LacZ/gland were delivered to both submandibular glands (SMGs) of NOD/LtJ mice at 8 weeks (before sialadenitis onset) of age. Salivary flow rates were determined at 8 weeks and time of killing. Blood glucose levels and body weights were measured weekly. After killing, saliva and SMGs were harvested. Analyses of salivary output, inflammatory infiltrates (focus score), SMG cytokine profile, body weight, and diabetes mellitus status were performed. Data from six different experimental studies over 2 years were analyzed and compared. RESULTS: Salivary flow rate, focus score, and SMG cytokines interleukin (IL)-2, IL-4, IL-6, IL-10, IL-12(p70), tumor necrosis factor-alpha and IFNgamma showed changes over time. There were no differences for body weight, diabetes mellitus prevalence, or blood glucose level of non-diabetic mice. CONCLUSION: This retrospective report is the first to describe longitudinal variability in the NOD mouse as a model for SS. We advise other investigators to continuously monitor the SS phenotype parameters and include appropriate controls when studying this disease in NOD mice.


Assuntos
Modelos Animais de Doenças , Síndrome de Sjogren/fisiopatologia , Análise de Variância , Animais , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Interferon gama/metabolismo , Interleucinas/metabolismo , Óperon Lac , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos NOD , Fenótipo , Saliva/metabolismo , Taxa Secretória , Síndrome de Sjogren/genética , Glândula Submandibular/imunologia , Glândula Submandibular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA