Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Obstet Gynecol ; 214(5): 631.e1-631.e11, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26874302

RESUMEN

BACKGROUND: The analysis of circulating microparticles in pregnancy is of revolutionary potential because it represents an in vivo biopsy of active gestational tissues. OBJECTIVE: We hypothesized that circulating microparticle signaling will differ in pregnancies that experience spontaneous preterm birth from those delivering at term and that these differences will be evident many weeks in advance of clinical presentation. STUDY DESIGN: Utilizing plasma specimens obtained between 10 and 12 weeks' gestation as part of a prospectively collected birth cohort in which pregnancy outcomes are independently validated by 2 board-certified maternal-fetal medicine physicians, 25 singleton cases of spontaneous preterm birth ≤ 34 weeks were matched by maternal age, race, and gestational age of sampling (±2 weeks) with 50 uncomplicated term deliveries. Circulating microparticles from these first-trimester specimens were isolated and analyzed by multiple reaction monitoring mass spectrometry for potential protein biomarkers following previous studies. Markers with robust univariate performance in correlating spontaneous preterm birth were further evaluated for their biological relevance via a combined functional profiling/pathway analysis and for multivariate performance. RESULTS: Among the 132 proteins evaluated, 62 demonstrated robust power of detecting spontaneous preterm birth in a bootstrap receiver-operating characteristic curve analysis at a false discovery rate of < 20% estimated via label permutation. Differential dependency network analysis identified spontaneous preterm birth-associated coexpression patterns linked to biological processes of inflammation, wound healing, and the coagulation cascade. Linear modeling of spontaneous preterm birth using a multiplex of the candidate biomarkers with a fixed sensitivity of 80% exhibited a specificity of 83% with median area under the curve of 0.89. These results indicate a strong potential of multivariate model development for informative risk stratification. CONCLUSION: This project has identified functional proteomic factors with associated biological processes that are already unique in their expression profiles at 10-12 weeks among women who go on to deliver spontaneously ≤ 34 weeks. These changes, with further validation, will allow the stratification of patients at risk of spontaneous preterm birth before clinical presentation.


Asunto(s)
Micropartículas Derivadas de Células , Nacimiento Prematuro/sangre , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Cromatografía Liquida , Estudios de Cohortes , Femenino , Humanos , Espectrometría de Masas , Embarazo , Primer Trimestre del Embarazo , Proteómica , Curva ROC , Medición de Riesgo , Sensibilidad y Especificidad
2.
Clin Immunol ; 146(3): 248-61, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23416241

RESUMEN

Cell-surface CD25 expression is critical for maintaining immune function and homeostasis. As in few reported cases, CD25 deficiency manifests with severe autoimmune enteritis and viral infections. To dissect the underlying immunological mechanisms driving these symptoms, we analyzed the regulatory and effector T cell functions in a CD25 deficient patient harboring a novel IL2RA mutation. Pronounced lymphoproliferation, mainly of the CD8(+) T cells, was detected together with an increase in T cell activation markers and elevated serum cytokines. However, Ag-specific responses were impaired in vivo and in vitro. Activated CD8(+)STAT5(+) T cells with lytic potential infiltrated the skin, even though FOXP3(+) Tregs were present and maintained a higher capacity to respond to IL-2 compared to other T-cell subsets. Thus, the complex pathogenesis of CD25 deficiency provides invaluable insight into the role of IL2/IL-2RA-dependent regulation in autoimmunity and inflammatory diseases.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Subunidad alfa del Receptor de Interleucina-2/deficiencia , Autoinmunidad/inmunología , Línea Celular , Proliferación Celular , Niño , Citocinas/sangre , Femenino , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/inmunología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Mutación , Piel/inmunología , Piel/patología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología
3.
J Immunol ; 186(6): 3779-86, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21317396

RESUMEN

IL-2 and TGF-ß1 play key roles in the immunobiology of Foxp3-expressing CD25(+)CD4(+) T cells (Foxp3(+)Treg). Administration of these cytokines offers an appealing approach to manipulate the Foxp3(+)Treg pool and treat T cell-mediated autoimmunity such as type 1 diabetes. However, efficacy of cytokine treatment is dependent on the mode of application, and the potent pleiotropic effects of cytokines like IL-2 may lead to severe side effects. In the current study, we used a gene therapy-based approach to assess the efficacy of recombinant adeno-associated virus vectors expressing inducible IL-2 or TGF-ß1 transgenes to suppress ongoing ß cell autoimmunity in NOD mice. Intramuscular vaccination of recombinant adeno-associated virus to 10-wk-old NOD female mice and a subsequent 3 wk induction of IL-2 was sufficient to prevent diabetes and block the progression of insulitis. Protection correlated with an increased frequency of Foxp3(+)Treg in the periphery as well as in the draining pancreatic lymph nodes and islets. IL-2 induced a shift in the ratio favoring Foxp3(+)Treg versus IFN-γ-expressing T cells infiltrating the islets. Induction of IL-2 had no systemic effect on the frequency or activational status of T cells and NK cells. Induction of TGF-ß1 had no effect on the Foxp3(+)Treg pool or the progression of ß cell autoimmunity despite induced systemic levels of activated TGF-ß1 that were comparable to IL-2. These results demonstrate that inducible IL-2 gene therapy is an effective and safe approach to manipulate Foxp3(+)Treg and suppress T cell-mediated autoimmunity and that under the conditions employed, IL-2 is more potent than TGF-ß1.


Asunto(s)
Dependovirus/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Terapia Genética/métodos , Interleucina-2/biosíntesis , Animales , Diabetes Mellitus Tipo 1/prevención & control , Femenino , Ingeniería Genética/métodos , Vectores Genéticos/inmunología , Vectores Genéticos/uso terapéutico , Células HEK293 , Humanos , Interleucina-2/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Infecciones por Parvoviridae/genética , Infecciones por Parvoviridae/inmunología , Infecciones por Parvoviridae/prevención & control , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Transgenes
4.
Eur J Immunol ; 41(5): 1480-1490, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21469091

RESUMEN

IL-2 plays a critical role in the induction and maintenance of FoxP3-expressing regulatory T cells (FoxP3(+) Tregs). Reduced expression of IL-2 is linked to T-cell-mediated autoimmune diseases such as type 1 diabetes (T1D), in which an imbalance between FoxP3(+) Tregs and pathogenic T effectors exists. We investigated the contribution of IL-2 to dysregulation of FoxP3(+) Tregs by comparing wildtype NOD mice with animals congenic for a C57BL/6-derived disease-resistant Il2 allele and in which T-cell secretion of IL-2 is increased (NOD.B6Idd3). Although NOD mice exhibited a progressive decline in the frequency of CD62L(hi) FoxP3(+) Tregs due to an increase in CD62L(lo) FoxP3(+) Tregs, CD62L(hi) FoxP3(+) Tregs were maintained in the pancreatic lymph nodes and islets of NOD.B6Idd3 mice. Notably, the frequency of proliferating CD62L(hi) FoxP3(+) Tregs was elevated in the islets of NOD.B6Idd3 versus NOD mice. Increasing levels of IL-2 in vivo also resulted in larger numbers of CD62L(hi) FoxP3(+) Tregs in NOD mice. These results demonstrate that IL-2 influences the suppressor activity of the FoxP3(+) Tregs pool by regulating the balance between CD62L(lo) and CD62L(hi) FoxP3(+) Tregs. In NOD mice, reduced IL-2 expression leads to an increase in nonsuppressive CD62L(lo) FoxP3(+) Tregs, which in turn correlates with a pool of CD62L(hi) FoxP3(+) Tregs with limited proliferation.


Asunto(s)
Interleucina-2/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Diabetes Mellitus Tipo 1/inmunología , Femenino , Factores de Transcripción Forkhead/análisis , Expresión Génica , Interleucina-2/genética , Interleucina-2/inmunología , Islotes Pancreáticos/inmunología , Selectina L/análisis , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Páncreas/inmunología
5.
Hepatology ; 53(5): 1696-707, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21520180

RESUMEN

UNLABELLED: Lentiviral vectors are attractive tools for liver-directed gene therapy because of their capacity for stable gene expression and the lack of preexisting immunity in most human subjects. However, the use of integrating vectors may raise some concerns about the potential risk of insertional mutagenesis. Here we investigated liver gene transfer by integrase-defective lentiviral vectors (IDLVs) containing an inactivating mutation in the integrase (D64V). Hepatocyte-targeted expression using IDLVs resulted in the sustained and robust induction of immune tolerance to both intracellular and secreted proteins, despite the reduced transgene expression levels in comparison with their integrase-competent vector counterparts. IDLV-mediated and hepatocyte-targeted coagulation factor IX (FIX) expression prevented the induction of neutralizing antibodies to FIX even after antigen rechallenge in hemophilia B mice and accounted for relatively prolonged therapeutic FIX expression levels. Upon the delivery of intracellular model antigens, hepatocyte-targeted IDLVs induced transgene-specific regulatory T cells that contributed to the observed immune tolerance. Deep sequencing of IDLV-transduced livers showed only rare genomic integrations that had no preference for gene coding regions and occurred mostly by a mechanism inconsistent with residual integrase activity. CONCLUSION: IDLVs provide an attractive platform for the tolerogenic expression of intracellular or secreted proteins in the liver with a substantially reduced risk of insertional mutagenesis.


Asunto(s)
Epítopos , Vectores Genéticos/genética , Hepatocitos , Tolerancia Inmunológica/genética , Integrasas/genética , Lentivirus/enzimología , Animales , Células Cultivadas , Daño del ADN , Femenino , Hepatocitos/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Riesgo
6.
Proc Natl Acad Sci U S A ; 106(26): 10770-4, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19541644

RESUMEN

The immune response has been implicated as a critical factor in determining the success or failure of clinical gene therapy trials. Generally, such a response is elicited by the desired transgene product or, in some cases, the delivery system. In the current study, we report the previously uncharacterized finding that a therapeutic cassette currently being used for human investigation displays alternative reading frames (ARFs) that generate unwanted protein products to induce a cytotoxic T lymphocyte (CTL) response. In particular, we tested the hypothesis that antigenic epitopes derived from an ARF in coagulation factor IX (F9) cDNA can induce CTL reactivity, subsequently killing F9-expressing hepatocytes. One peptide (p18) of 3 candidates from an ARF of the F9 transgene induced CD8(+) T cell reactivity in mice expressing the human MHC class I molecule B0702. Subsequently, upon systemic administration of adeno-associated virus (AAV) serotype 2 vectors packaged with the F9 transgene (AAV2/F9), a robust CD8(+) CTL response was elicited against peptide p18. Of particular importance is that the ARF epitope-specific CTLs eliminated AAV2/F9-transduced hepatocytes but not AAV2/F9 codon-optimized (AAV2/F9-opt)-transduced liver cells in which p18 epitope was deleted. These results demonstrate a previously undiscovered mechanism by which CTL responses can be elicited by cryptic epitopes generated from a therapeutic transgene and have significant implications for all gene therapy modalities. Such unforeseen epitope generation warrants careful analysis of transgene sequences for ARFs to reduce the potential for adverse events arising from immune responses during clinical gene therapy protocols.


Asunto(s)
Epítopos/inmunología , Factor IX/metabolismo , Terapia Genética/métodos , Inmunidad Celular/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células COS , Chlorocebus aethiops , Técnicas de Cocultivo , Citotoxicidad Inmunológica/inmunología , Dependovirus/genética , Epítopos/genética , Factor IX/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Antígenos HLA-B/genética , Antígenos HLA-B/metabolismo , Humanos , Hibridomas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligopéptidos/inmunología , Sistemas de Lectura Abierta/genética , Bazo/citología , Bazo/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Transfección
7.
J Virol ; 83(13): 6817-24, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19369348

RESUMEN

A recent clinical trial in patients with hemophilia B has suggested that adeno-associated virus (AAV) capsid-specific cytotoxic T lymphocytes (CTLs) eliminated AAV-transduced hepatocytes and resulted in therapeutic failure. AAV capsids elicit a CTL response in animal models; however, these capsid-specific CTLs fail to kill AAV-transduced target cells in mice. To better model the human clinical trial data in mice, we introduced an immunodominant epitope derived from ovalbumin (OVA; SIINFEKL) into the AAV capsid and tested CTL-mediated killing of AAV2-transduced target tissues in vivo. Initially, in vitro experiments demonstrated both classical class I and cross-presentation of the OVA antigen, following endogenous expression or AAV2-OVA vector transduction, respectively. Furthermore, an OVA-specific CTL response was elicited after muscular or systemic injection of the AAV2-OVA vector. Finally, CTL reactivity was enhanced in mice with established SIINFEKL-specific immunity after AAV2-OVA/alpha1 anti-trypsin (AAT) administration. Most importantly, these OVA-specific CTLs decreased AAT expression in mice treated with AAV2-OVA/AAT vector that followed a time course mimicking uncoating kinetics of AAV2 transduction in OVA-immunized mice. These results demonstrate that AAV capsid-derived antigens elicit CD8(+) CTL reactivity, and these CTLs eliminated AAV-transduced target cells in mice. Notably, this model system can be exploited to study the kinetics of capsid presentation from different serotypes of AAV and permit the design of novel strategies to block CTL-mediated killing of AAV-transduced cells.


Asunto(s)
Dependovirus/inmunología , Vectores Genéticos/inmunología , Epítopos Inmunodominantes/inmunología , Ovalbúmina/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos Virales/inmunología , Cápside/inmunología , Línea Celular , Dependovirus/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/virología , Transfección
8.
Clin Immunol ; 129(1): 49-57, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18675592

RESUMEN

Type 1 diabetes (T1D) is characterized by the T cell mediated destruction of the insulin-producing beta cells. Antigen-specific immunotherapies are used to selectively tolerize beta cell-specific pathogenic T cells either directly, or indirectly through the induction of immunoregulatory T cells. A key concern of antigen-specific immunotherapy is exacerbating autoimmunity. We compared the T cell reactivity and efficacy induced by plasmid DNA (pDNA) encoding glutamic acid decarboxylase 65 (GAD65) administered via intramuscular versus gene gun vaccination in NOD mice at a late preclinical stage of T1D. Whereas intramuscular injection of pGAD65 promoted a predominant type 1 CD4(+) T cell response and failed to suppress ongoing beta cell autoimmunity, gene gun vaccination preferentially induced IL-4 secreting CD4(+) T cells and significantly delayed the onset of diabetes. These findings demonstrate that gene gun delivery of autoantigen-encoding pDNA preferentially elicits immunoregulatory T cells and offers a safe, effective mode of pDNA vaccination for the treatment of T1D and other autoimmune diseases.


Asunto(s)
Biolística , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Glutamato Descarboxilasa/genética , Linfocitos T Reguladores/inmunología , Vacunas de ADN/administración & dosificación , Animales , Autoantígenos/inmunología , Autoinmunidad , Diabetes Mellitus Tipo 1/genética , Glutamato Descarboxilasa/sangre , Glutamato Descarboxilasa/inmunología , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Células Secretoras de Insulina/inmunología , Interleucina-4/inmunología , Ratones , Ratones Endogámicos NOD , Proteínas Recombinantes de Fusión/sangre , Linfocitos T Reguladores/metabolismo , Transgenes , Vacunación , Vacunas de ADN/inmunología
9.
Diabetes ; 52(3): 708-16, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12606512

RESUMEN

Islet transplantation represents a potential cure for type 1 diabetes, yet persistent autoimmune and allogeneic immunities currently limit its clinical efficacy. For alleviating the autoimmune destruction of transplanted islets, newly diagnosed NOD mice were provided a single intramuscular injection of recombinant adeno-associated viral vector encoding murine IL-10 (rAAV-IL-10) 4 weeks before renal capsule delivery of 650 syngeneic islets. A dose-dependent protection of islet grafts was observed. Sixty percent (3 of 5) of NOD mice that received a transduction of a high-dose (4 x 10(9) infectious units) rAAV-IL-10 remained normoglycemic for at least 117 days, whereas diabetes recurred within 17 days in mice that received a low-dose rAAV-IL-10 (4 x 10(8) infectious units; 5 of 5) as well as in all of the control mice (5 of 5 untreated and 4 of 4 rAAV-green fluorescent protein-transduced). Serum IL-10 levels positively correlated with prolonged graft survival and were negatively associated with the intensity of autoimmunity. The mechanism of rAAV-IL-10 protection involved a reduction of lymphocytic infiltration as well as induction of antioxidant enzymes manganese superoxide dismutase and heme oxygenase 1 in islet grafts. These studies support the utility of immunoregulatory cytokine gene therapy delivered by rAAV for preventing autoimmune disease recurrence in transplant-based therapies for type 1 diabetes.


Asunto(s)
Dependovirus/genética , Diabetes Mellitus Tipo 1/terapia , Terapia Genética , Interleucina-10/genética , Trasplante de Islotes Pancreáticos , Prevención Secundaria , Animales , Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Expresión Génica , Vectores Genéticos , Supervivencia de Injerto , Proteínas Fluorescentes Verdes , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo-Oxigenasa 1 , Inflamación/patología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Proteínas Luminiscentes/genética , Linfocitos/patología , Proteínas de la Membrana , Ratones , Ratones Endogámicos NOD , Músculo Esquelético/metabolismo , Superóxido Dismutasa/biosíntesis
10.
Int Rev Immunol ; 24(5-6): 307-26, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16318984

RESUMEN

A major effort has been on-going to develop immunotherapies to prevent and/or treat type 1 diabetes (T1D). This autoimmune disease is characterized by the selective loss of the insulin-producing beta cells via the cumulative effects of autoantigen-specific CD4(+) and CD8(+) T cells, autoantibodies, and activated antigen-presenting cells. To be applicable in a clinical setting, immunotherapies must suppress established beta-cell autoimmunity. Preclinical studies and recent clinical findings suggest that antigen-specific and systemic-based strategies can be effective in this regard. However, either approach alone may not be sufficient to block the diabetogenic response and establish long-term protection in the clinic. In this review, we will discuss the importance of both strategies and how a combinatorial approach to treat T1D is appealing.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Inmunoterapia/métodos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Complejo CD3/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Terapia Genética/métodos , Humanos , Terapia de Inmunosupresión/métodos , Células Secretoras de Insulina/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
11.
Sci Transl Med ; 7(289): 289ra81, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26019217

RESUMEN

Antigen (Ag)-specific tolerance in type 1 diabetes (T1D) in human has not been achieved yet. Targeting lentiviral vector (LV)-mediated gene expression to hepatocytes induces active tolerance toward the encoded Ag. The insulin B chain 9-23 (InsB9-23) is an immunodominant T cell epitope in nonobese diabetic (NOD) mice. To determine whether auto-Ag gene transfer to hepatocytes induces tolerance and control of T1D, NOD mice were treated with integrase-competent LVs (ICLVs) that selectively target the expression of InsB9-23 to hepatocytes. ICLV treatment induced InsB9-23-specific effector T cells but also FoxP3(+) regulatory T cells (Tregs), which halted islet immune cell infiltration, and protected from T1D. Moreover, ICLV treatment combined with a single suboptimal dose of anti-CD3 monoclonal antibody (mAb) is effective in T1D reversal. Splenocytes from LV.InsB9-23-treated mice, but not from LV.OVA (ovalbumin)-treated control mice, stopped diabetes development, demonstrating that protection is Ag-specific. Depletion of CD4(+)CD25(+)FoxP3(+) T cells led to diabetes progression, indicating that Ag-specific FoxP3(+) Tregs mediate protection. Integrase-defective LVs (IDLVs).InsB9-23, which alleviate the concerns for insertional mutagenesis and support transient transgene expression in hepatocytes, were also efficient in protecting from T1D. These data demonstrate that hepatocyte-targeted auto-Ag gene expression prevents and resolves T1D and that stable integration of the transgene is not required for this protection. Gene transfer to hepatocytes can be used to induce Ag-specific tolerance in autoimmune diseases.


Asunto(s)
Antígenos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Factores de Transcripción Forkhead/metabolismo , Técnicas de Transferencia de Gen , Hepatocitos/metabolismo , Insulina/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Linfocitos T Reguladores/inmunología , Animales , Diabetes Mellitus Tipo 1/prevención & control , Progresión de la Enfermedad , Vectores Genéticos/metabolismo , Tolerancia Inmunológica , Insulina/genética , Integrasas/metabolismo , Lentivirus/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Fragmentos de Péptidos/genética , Linfocitos T Citotóxicos/inmunología , Transgenes
12.
Ann N Y Acad Sci ; 1005: 237-41, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14679068

RESUMEN

The concept of insulin production afforded by hepatic gene therapy retains promise as a potential therapy for type 1 diabetes, but the approach has been limited by the need for strict transgene regulation in response to fluctuating levels of both glucose and insulin. Furthermore, while hepatocytes contain various glucose-responsive elements, they lack the appropriate regulated secretory system necessary for insulin release, thereby necessitating the requirement for transcriptional regulation of hepatic insulin production under the direction of a glucose-responsive promoter. To address this, we have evaluated several glucose-responsive promoters that may be used successfully for hepatic insulin production via recombinant adeno-associated virus (rAAV) therapy. Our results suggest that the human insulin promoter represents a strong candidate as a robust, glucose-responsive promoter for regulated hepatic insulin production.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Glucosa/fisiología , Insulina/genética , Neoplasias Hepáticas Experimentales/metabolismo , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Carcinoma Hepatocelular/patología , Cartilla de ADN , Fructosa-Bifosfatasa/genética , Glucosa-6-Fosfatasa/genética , Humanos , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Endogámicos NOD , Ratas , Células Tumorales Cultivadas
13.
Transl Res ; 161(4): 230-40, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23360745

RESUMEN

The use of lentiviral vectors (LV)s for in vivo gene therapy is an ideal platform for treating many types of disease. Since LVs can transduce a wide array of cells, support long-term gene expression, and be modified to enhance cell targeting, LVs are a powerful modality to deliver life-long therapeutic proteins. A major limitation facing the use of LVs for in vivo gene therapy is the induction of immune responses, which can reduce the transduction efficiency of LV, eliminate the transduced cells, and inhibit the effect of the therapeutic protein. LV strategies designed to restrict transgene expression to the liver to exploit its naturally tolerogenic properties have proven to significantly reduce the induction of pathogenic immune responses and increase therapeutic efficacy. In this review, we outline the immunological hurdles facing in vivo LV gene therapy and highlight the advantages and limitations of using liver-directed LV gene therapy.


Asunto(s)
Terapia Genética/métodos , Inmunidad/inmunología , Lentivirus/genética , Hígado/inmunología , Animales , Humanos , Tolerancia Inmunológica/inmunología , Transgenes/genética
14.
EMBO Mol Med ; 5(11): 1684-97, 2013 11.
Artículo en Inglés | MEDLINE | ID: mdl-24106222

RESUMEN

A major complication of factor replacement therapy for haemophilia is the development of anti-factor neutralizing antibodies (inhibitors). Here we show that liver gene therapy by lentiviral vectors (LVs) expressing factor IX (FIX) strongly reduces pre-existing anti-FIX antibodies and eradicates FIX inhibitors in haemophilia B mice. Concomitantly, plasma FIX levels and clotting activity rose to 50-100% of normal. The treatment was effective in 75% of treated mice. FIX-specific plasma cells (PCs) and memory B cells were reduced, likely because of memory B-cell depletion in response to constant exposure to high doses of FIX. Regulatory T cells displaying FIX-specific suppressive capacity were induced in gene therapy treated mice and controlled FIX-specific T helper cells. Gene therapy proved safer than a regimen mimicking immune tolerance induction (ITI) by repeated high-dose FIX protein administration, which induced severe anaphylactoid reactions in inhibitors-positive haemophilia B mice. Liver gene therapy can thus reverse pre-existing immunity, induce active tolerance to FIX and establish sustained FIX activity at therapeutic levels. These data position gene therapy as an attractive treatment option for inhibitors-positive haemophilic patients.


Asunto(s)
Factor IX/genética , Terapia Genética , Hemofilia B/genética , Hemofilia B/terapia , Lentivirus/fisiología , Hígado/virología , Animales , Anticuerpos/inmunología , Linfocitos B/inmunología , Línea Celular , Factor IX/inmunología , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Hemofilia B/inmunología , Humanos , Lentivirus/genética , Hígado/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Front Immunol ; 3: 30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22566914

RESUMEN

The immuno-regulatory mechanisms of IL-10-producing type 1 regulatory T (Tr1) cells have been widely studied over the years. However, several recent discoveries have shed new light on the cellular and molecular mechanisms that human Tr1 cells use to control immune responses and induce tolerance. In this review we outline the well known and newly discovered regulatory properties of human Tr1 cells and provide an in-depth comparison of the known suppressor mechanisms of Tr1 cells with FOXP3(+) T(reg). We also highlight the role that Tr1 cells play in promoting and maintaining tolerance in autoimmunity, allergy, and transplantation.

16.
Front Microbiol ; 2: 221, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22144977

RESUMEN

The success of in vivo gene therapy greatly depends on the ability to control the immune response toward the therapeutic transgene. Over the last decade several vector-based and pharmacological approaches have been explored to control the immune-mediated clearance of transgene-expressing cells after viral delivery. One important outcome from these studies is the concept that expression of a transgene in tolerance-promoting organs, such as the liver and tolerogenic antigen-presenting cells, can help safeguard transgene-expressing cells from immune-mediated clearance. Gene therapists are now manipulating vectors to target naturally occurring tolerogenic properties of the body by: (i) incorporating tissue/cell specific promoters for targeted expression, (ii) using viral-capsid engineering to alter tropism and avoid pre-existing immunity, and (iii) regulating cell and activation dependent expression by including micro-RNA (miR) targets into expression cassettes. The combination of these three layers of vector regulation greatly enhances the targeting of tolerogenic cells and limits off-target expression of the transgene, which can lead to the induction of transgene-specific pathogenic effector T cells. In this review, we discuss the application of using miR transgene regulation to generate tolerogenic responses and speculate on possible mechanisms used by the liver to induce the transgene-specific regulatory T cells.

17.
Diabetes ; 56(5): 1395-402, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17317763

RESUMEN

Antigen-specific immunotherapy, an approach to selectively block autoimmune diabetes, generally declines in nonobese diabetic (NOD) mice as disease progresses. To define the parameters influencing the efficacy of antigen-specific immunotherapy once diabetes is established, plasmid DNA (pDNA) vaccination was used to suppress autoimmune-mediated destruction of syngeneic islet grafts in diabetic NOD recipients. pDNAs encoding a glutamic acid decarboxylase 65 (GAD65)-Ig molecule (pGAD65), interleukin (IL)-4 (pIL4), and IL-10 (pIL10) significantly delayed the onset of recurrent diabetes compared with pGAD65+pIL10-vaccinated recipients. Despite differences in efficacy, a similar frequency of GAD65-specific CD4(+) T-cells secreting IL-4, IL-10, or interferon-gamma were detected in mice treated with pGAD65+pIL4+pIL10 and pGAD65+pIL10. However, the frequency of FoxP3-expressing CD4(+)CD25(+)CD62L(hi) T-cells was increased in the renal and pancreatic lymph nodes of diabetic recipients vaccinated with pGAD65+pIL4+pIL10. These immunoregulatory CD4(+)CD25(+) T-cells (CD4(+)CD25(+) Treg) exhibited enhanced in vivo and in vitro suppressor activity that partially was transforming growth factor-beta dependent. Furthermore, duration of islet graft protection in pGAD65+pIL4+pIL10-vaccinated diabetic recipients correlated with the persistence of CD4(+)CD25(+) Treg. These data demonstrate that the frequency and maintenance of FoxP3-expressing CD4(+)CD25(+) Treg influence antigen-induced suppression of ongoing beta-cell autoimmunity in diabetic recipients.


Asunto(s)
Antígenos/inmunología , Antígenos/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Inmunoterapia/métodos , Animales , Recuento de Linfocito CD4 , Diabetes Mellitus Tipo 1/terapia , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glutamato Descarboxilasa/inmunología , Células Secretoras de Insulina/inmunología , Subunidad alfa del Receptor de Interleucina-2/deficiencia , Subunidad alfa del Receptor de Interleucina-2/genética , Interleucina-4/inmunología , Isoenzimas/inmunología , Ratones , Ratones Endogámicos NOD , Muramidasa/inmunología , Reacción en Cadena de la Polimerasa , Factor de Crecimiento Transformador beta/análisis
18.
J Immunol ; 178(3): 1388-95, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17237386

RESUMEN

A major issue regarding T cell responses in autoimmunity is how the repertoire compares between the periphery and target organ. In type 1 diabetes, the status of at-risk or diabetic individuals can be monitored by measuring beta cell-specific T cells isolated from PBL, but whether these T cells accurately reflect the repertoire residing in the pancreatic islets is unclear. The TCR repertoire of disease-relevant, tetramer-sorted CD8(+) T cells was examined at the single-cell level in PBL, pancreatic lymph nodes (PLN), and the islets of individual NOD mice. CDR3alpha and CDR3beta sequences demonstrated that the same repertoire of T cells in PBL was detected in the islets and PLN, although the frequency of specific clonotypes varied. Albeit infrequent, clonotypes that were prevalent in the islets but not found in PBL were also detected. beta cell Ag immunization expanded immunodominant PBL clonotypes present in the islets and PLN. These results show that insight into repertoire profiles of islet-infiltrating T cells can be obtained from PBL.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Secretoras de Insulina/inmunología , Islotes Pancreáticos/citología , Especificidad del Receptor de Antígeno de Linfocitos T , Animales , Autoinmunidad , Células Sanguíneas/citología , Movimiento Celular , Células Clonales/inmunología , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos NOD
19.
J Immunol ; 171(5): 2270-8, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12928371

RESUMEN

Early systemic treatment of nonobese diabetic mice with high doses of recombinant adeno-associated virus (rAAV) vector expressing murine IL-10 prevents type 1 diabetes. To determine the therapeutic parameters and immunological mechanisms underlying this observation, female nonobese diabetic mice at 4, 8, and 12 wk of age were given a single i.m. injection of rAAV-murine IL-10 (10(4), 10(6), 10(8), and 10(9) infectious units (IU)), rAAV-vector expressing truncated murine IL-10 fragment (10(9) IU), or saline. Transduction with rAAV-IL-10 at 10(9) IU completely prevented diabetes in all animals injected at all time points, including, surprisingly, 12-wk-old animals. Treatment with 10(8) IU provided no protection in the 12-wk-old injected mice, partial prevention in 8-wk-old mice, and full protection in all animals injected at 4 wk of age. All other treatment groups developed diabetes at a similar rate. The rAAV-IL-10 therapy attenuated pancreatic insulitis, decreased MHC II expression on CD11b+ cells, increased the population of CD11b+ cells, and modulated insulin autoantibody production. Interestingly, rAAV-IL-10 therapy dramatically increased the percentage of CD4+CD25+ regulatory T cells. Adoptive transfer studies suggest that rAAV-IL-10 treatment alters the capacity of splenocytes to impart type 1 diabetes in recipient animals. This study indicates the potential for immunomodulatory gene therapy to prevent autoimmune diseases, including type 1 diabetes, and implicates IL-10 as a molecule capable of increasing the percentages of regulatory cells in vivo.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Terapia Genética/métodos , Interleucina-10/genética , Interleucina-10/uso terapéutico , Receptores de Interleucina-2/biosíntesis , Subgrupos de Linfocitos T/inmunología , Adenoviridae/genética , Animales , Formación de Anticuerpos/genética , Autoanticuerpos/biosíntesis , Linfocitos T CD4-Positivos/metabolismo , Dependovirus/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Relación Dosis-Respuesta Inmunológica , Femenino , Vectores Genéticos , Inmunidad Celular/genética , Inyecciones Intramusculares , Anticuerpos Insulínicos/biosíntesis , Interleucina-10/biosíntesis , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Recombinación Genética , Subgrupos de Linfocitos T/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA