Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetologia ; 67(1): 27-41, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37782353

RESUMEN

AIMS/HYPOTHESIS: We hypothesised that islet beta cell antigen presentation in the gut along with a tolerising cytokine would lead to antigen-specific tolerance in type 1 diabetes. We evaluated this in a parallel open-label Phase 1b study using oral AG019, food-grade Lactococcus lactis bacteria genetically modified to express human proinsulin and human IL-10, as a monotherapy and in a parallel, randomised, double-blind Phase 2a study using AG019 in combination with teplizumab. METHODS: Adults (18-42 years) and adolescents (12-17 years) with type 1 diabetes diagnosed within 150 days were enrolled, with documented evidence of at least one autoantibody and a stimulated peak C-peptide level >0.2 nmol/l. Participants were allocated to interventions using interactive response technology. We treated 42 people aged 12-42 years with recent-onset type 1 diabetes, 24 with Phase 1b monotherapy (open-label) and 18 with Phase 2a combination therapy. In the Phase 2a study, after treatment of the first two open-label participants, all people involved were blinded to group assignment, except for the Data Safety Monitoring Board members and the unblinded statistician. The primary endpoint was safety and tolerability based on the incidence of treatment-emergent adverse events, collected up to 6 months post treatment initiation. The secondary endpoints were pharmacokinetics, based on AG019 detection in blood and faeces, and pharmacodynamic activity. Metabolic and immune endpoints included stimulated C-peptide levels during a mixed meal tolerance test, HbA1c levels, insulin use, and antigen-specific CD4+ and CD8+ T cell responses using an activation-induced marker assay and pooled tetramers, respectively. RESULTS: Data from 24 Phase 1b participants and 18 Phase 2a participants were analysed. No serious adverse events were reported and none of the participants discontinued AG019 due to treatment-emergent adverse events. No systemic exposure to AG019 bacteria, proinsulin or human IL-10 was demonstrated. In AG019 monotherapy-treated adults, metabolic variables were stabilised up to 6 months (C-peptide, insulin use) or 12 months (HbA1c) post treatment initiation. In participants treated with AG019/teplizumab combination therapy, all measured metabolic variables stabilised or improved up to 12 months and CD8+ T cells with a partially exhausted phenotype were significantly increased at 6 months. Circulating preproinsulin-specific CD4+ and CD8+ T cells were detected before and after treatment, with a reduction in the frequency of preproinsulin-specific CD8+ T cells after treatment with monotherapy or combination therapy. CONCLUSIONS/INTERPRETATION: Oral delivery of AG019 was well tolerated and safe as monotherapy and in combination with teplizumab. AG019 was not shown to interfere with the safety profile of teplizumab and may have additional biological effects, including changes in preproinsulin-specific T cells. These preliminary data support continuing studies with this agent alone and in combination with teplizumab or other systemic immunotherapies in type 1 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT03751007, EudraCT 2017-002871-24 FUNDING: This study was funded by Precigen ActoBio.


Asunto(s)
Diabetes Mellitus Tipo 1 , Adulto , Adolescente , Humanos , Interleucina-10 , Péptido C , Linfocitos T CD8-positivos/metabolismo , Proinsulina , Método Doble Ciego
2.
Diabetes ; 72(10): 1470-1482, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37494666

RESUMEN

Immunomodulation combined with antigen therapy holds great promise to arrest autoimmune type 1 diabetes, but clinical translation is hampered by a lack of prognostic biomarkers. Low-dose anti-CD3 plus Lactococcus lactis bacteria secreting proinsulin and IL-10 reversed new-onset disease in nonobese diabetic (NOD) mice, yet some mice were resistant to the therapy. Using miRNA profiling, six miRNAs (i.e., miR-34a-5p, miR-125a-3p, miR-193b-3p, miR-328, miR-365-3p, and miR-671-3p) were identified as differentially expressed in plasma of responder versus nonresponder mice before study entry. After validation and stratification in an independent cohort, plasma miR-193b-3p and miR-365-3p, combined with age and glycemic status at study entry, had the best power to predict, with high sensitivity and specificity, poor response to the therapy. These miRNAs were highly abundant in pancreas-infiltrating neutrophils and basophils with a proinflammatory and activated phenotype. Here, a set of miRNAs and disease-associated parameters are presented as a predictive signature for the L. lactis-based immunotherapy outcome in new-onset type 1 diabetes, hence allowing targeted recruitment of trial participants and accelerated trial execution. ARTICLE HIGHLIGHTS: Low-dose anti-CD3 combined with oral gavage of genetically modified Lactococcus lactis bacteria secreting human proinsulin and IL-10 holds great promise to arrest autoimmune type 1 diabetes, but the absence of biomarkers predicting therapeutic success hampers clinical translation. A set of cell-free circulation miRNAs together with age and glycemia at baseline predicts a poor response after L. lactis-based immunotherapy in nonobese mice with new-onset diabetes. Pancreas-infiltrating neutrophils and basophils are identified as potential cellular sources of discovered miRNAs. The prognostic signature could guide targeted recruitment of patients with newly diagnosed type 1 diabetes in clinical trials with the L. lactis-based immunotherapy.


Asunto(s)
Diabetes Mellitus Tipo 1 , Lactococcus lactis , MicroARNs , Humanos , Animales , Ratones , Diabetes Mellitus Tipo 1/terapia , Interleucina-10 , Lactococcus lactis/genética , Proinsulina/genética , Perfilación de la Expresión Génica , MicroARNs/genética , Biomarcadores , Ratones Endogámicos NOD , Inmunoterapia
3.
Front Immunol ; 11: 1103, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582188

RESUMEN

A combination treatment (CT) of proinsulin and IL-10 orally delivered via genetically modified Lactococcus lactis bacteria combined with low-dose anti-CD3 (aCD3) therapy successfully restores glucose homeostasis in newly diagnosed non-obese diabetic (NOD) mice. Tolerance is accompanied by the accumulation of Foxp3+ regulatory T cells (Tregs) in the pancreas. To test the potential of this therapy outside the window of acute diabetes diagnosis, we substituted autoimmune diabetic mice, with disease duration varying between 4 and 53 days, with syngeneic islets at the time of therapy initiation. Untreated islet recipients consistently showed disease recurrence after 8.2 ± 0.7 days, while 32% of aCD3-treated and 48% of CT-treated mice remained normoglycemic until 6 weeks after therapy initiation (P < 0.001 vs. untreated controls for both treatments, P < 0.05 CT vs. aCD3 therapy). However, mice that were diabetic for more than 2 weeks before treatment initiation were less efficient at maintaining normoglycemia than those treated within 2 weeks of diabetes diagnosis, particularly in the aCD3-treated group. The complete elimination of endogenous beta cell mass with alloxan at the time of diabetes diagnosis pointed toward the significance of continuous feeding of the islet antigen proinsulin at the time of aCD3 therapy for treatment success. The CT providing proinsulin protected 69% of mice, compared to 33% when an irrelevant antigen (ovalbumin) was combined with aCD3 therapy, or to 27% with aCD3 therapy alone. Sustained tolerance was accompanied with a reduction of IGRP+CD8+ autoreactive T cells and an increase in insulin-reactive (InsB12-20 or InsB13-2) Foxp3+CD4+ Tregs, with a specific accumulation of Foxp3+ Tregs around the insulin-containing islet grafts after CT with proinsulin. The combination of proinsulin and IL-10 via oral Lactococcus lactis with low-dose aCD3 therapy can restore tolerance to beta cells in autoimmune diabetic mice, also when therapy is started outside the window of acute diabetes diagnosis, providing persistence of insulin-containing islets or prolonged beta cell function.


Asunto(s)
Complejo CD3/antagonistas & inhibidores , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/efectos de los fármacos , Interleucina-10/administración & dosificación , Proinsulina/administración & dosificación , Animales , Diabetes Mellitus Experimental/inmunología , Vectores Genéticos , Humanos , Lactococcus lactis , Ratones , Ratones Endogámicos NOD , Autotolerancia/efectos de los fármacos , Autotolerancia/inmunología
4.
Diabetes ; 66(2): 448-459, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28108611

RESUMEN

The introduction of ß-cell autoantigens via the gut through Lactococcus lactis (L. lactis) has been demonstrated to be a promising approach for diabetes reversal in NOD mice. Here we show that a combination therapy of low-dose anti-CD3 with a clinical-grade self-containing L. lactis, appropriate for human application, secreting human proinsulin and interleukin-10, cured 66% of mice with new-onset diabetes, which is comparable to therapy results with plasmid-driven L. lactis Initial blood glucose concentrations (<350 mg/dL) and insulin autoantibody positivity were predictors of the stable reversal of hyperglycemia, and decline in insulin autoantibody positivity was an immune biomarker of therapeutic outcome. The assessment of the immune changes induced by the L. lactis-based therapy revealed elevated frequencies of CD4+Foxp3+ T cells in the pancreas-draining lymph nodes, pancreas, and peripheral blood of all treated mice, independent of metabolic outcome. Neutralization of cytotoxic T-lymphocyte antigen 4 and transforming growth factor-ß partially abrogated the suppressive function of therapy-induced regulatory T cells (Tregs). Ablation or functional impairment of Foxp3+ Tregs in vivo at the start or stop of therapy impaired immune tolerance, highlighting the dependence of the therapy-induced tolerance in mice with new-onset diabetes on the presence and functionality of CD4+Foxp3+ T cells. Biomarkers identified in this study can potentially be used in the future to tailor the L. lactis-based combination therapy for individual patients.


Asunto(s)
Anticuerpos/farmacología , Glucemia/efectos de los fármacos , Diabetes Mellitus/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Interleucina-10/metabolismo , Lactobacillus/metabolismo , Proinsulina/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Animales , Anticuerpos Neutralizantes/farmacología , Glucemia/metabolismo , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Antígeno CTLA-4/efectos de los fármacos , Antígeno CTLA-4/inmunología , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/inmunología , Prueba de Tolerancia a la Glucosa , Tolerancia Inmunológica/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Páncreas/efectos de los fármacos , Páncreas/patología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/inmunología
5.
Diabetes ; 63(8): 2876-87, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24677716

RESUMEN

Growing insight into the pathogenesis of type 1 diabetes (T1D) and numerous studies in preclinical models highlight the potential of antigen-specific approaches to restore tolerance efficiently and safely. Oral administration of protein antigens is a preferred method for tolerance induction, but degradation during gastrointestinal passage can impede such protein-based therapies, reducing their efficacy and making them cost-ineffective. To overcome these limitations, we generated a tolerogenic bacterial delivery technology based on live Lactococcus lactis (LL) bacteria for controlled secretion of the T1D autoantigen GAD65370-575 and the anti-inflammatory cytokine interleukin-10 in the gut. In combination with short-course low-dose anti-CD3, this treatment stabilized insulitis, preserved functional ß-cell mass, and restored normoglycemia in recent-onset NOD mice, even when hyperglycemia was severe at diagnosis. Combination therapy did not eliminate pathogenic effector T cells, but increased the presence of functional CD4(+)Foxp3(+)CD25(+) regulatory T cells. These preclinical data indicate a great therapeutic potential of orally administered autoantigen-secreting LL for tolerance induction in T1D.


Asunto(s)
Autoantígenos/farmacología , Diabetes Mellitus/inmunología , Glutamato Descarboxilasa/farmacología , Interleucina-10/metabolismo , Fragmentos de Péptidos/farmacología , Administración Oral , Envejecimiento , Animales , Autoantígenos/administración & dosificación , Autoantígenos/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Glutamato Descarboxilasa/administración & dosificación , Interleucina-10/genética , Lactococcus lactis , Ratones , Ratones Endogámicos NOD , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Linfocitos T Reguladores/efectos de los fármacos
6.
J Clin Invest ; 122(5): 1717-25, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22484814

RESUMEN

Current interventions for arresting autoimmune diabetes have yet to strike the balance between sufficient efficacy, minimal side effects, and lack of generalized immunosuppression. Introduction of antigen via the gut represents an appealing method for induction of antigen-specific tolerance. Here, we developed a strategy for tolerance restoration using mucosal delivery in mice of biologically contained Lactococcus lactis genetically modified to secrete the whole proinsulin autoantigen along with the immunomodulatory cytokine IL-10. We show that combination therapy with low-dose systemic anti-CD3 stably reverted diabetes in NOD mice and increased frequencies of local Tregs, which not only accumulated in the pancreatic islets, but also suppressed immune response in an autoantigen-specific way. Cured mice remained responsive to disease-unrelated antigens, which argues against excessive immunosuppression. Application of this therapeutic tool achieved gut mucosal delivery of a diabetes-relevant autoantigen and a biologically active immunomodulatory cytokine, IL-10, and, when combined with a low dose of systemic anti-CD3, was well tolerated and induced autoantigen-specific long-term tolerance, allowing reversal of established autoimmune diabetes. Therefore, we believe this method could be an effective treatment strategy for type 1 diabetes in humans.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Tolerancia Inmunológica , Lactococcus lactis/genética , Animales , Autoantígenos/biosíntesis , Autoantígenos/genética , Complejo CD3/inmunología , Recuento de Células , Proliferación Celular , Terapia Combinada , Diabetes Mellitus Tipo 1/inmunología , Humanos , Hipoglucemiantes/uso terapéutico , Factores Inmunológicos/uso terapéutico , Terapia de Inmunosupresión , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Interleucina-10/biosíntesis , Interleucina-10/genética , Interleucina-10/metabolismo , Mucosa Intestinal , Lactococcus lactis/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Proinsulina/biosíntesis , Proinsulina/genética , Proinsulina/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/fisiología
7.
Oral Oncol ; 46(7): 564-70, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20542722

RESUMEN

Non-clinical studies, focusing on the pharmacodynamics (PD), pharmacokinetics (PK) and safety pharmacology of genetically modified Lactococcus lactis (L. lactis) bacteria, engineered to secrete human Trefoil Factor 1 (hTFF1), were performed to provide proof-of-concept for the treatment of oral mucositis (OM) patients. L. lactis strain sAGX0085 was constructed by stably inserting an htff1 expression cassette into the bacterial genome, and clinically formulated as a mouth rinse (coded AG013). PD studies, using different oral dosing regimens, were performed in a clinically relevant hamster model for radiation-induced OM. The PK profile was assessed in healthy hamsters and in hamsters with radiation-induced OM. In addition, in vitro and in vivo safety pharmacology studies were conducted, in pooled, complement-preserved human serum, and in neutropenic hamsters and rats respectively. Topical administration of L. lactis sAGX0085/AG013 to the oral mucosa significantly reduced the severity and course of radiation-induced OM. PK studies demonstrated that both living L. lactis bacteria, as well as the hTFF1 secreted, could be recovered from the administration site for maximum 24h post-dosing, without systemic exposure. The in vitro and in vivo safety pharmacology studies confirmed that L. lactis sAGX0085 could not survive in systemic circulation, not even under neutropenic conditions. The results from the PD, PK and safety pharmacology studies reported here indicate that in situ secretion of hTFF1 by topically administered L. lactis bacteria provides a safe and efficacious therapeutic tool for the prevention and treatment of OM.


Asunto(s)
Lactococcus lactis/metabolismo , Antisépticos Bucales/metabolismo , Péptidos/metabolismo , Estomatitis/tratamiento farmacológico , Animales , Cricetinae , Humanos , Antisépticos Bucales/farmacocinética , Péptidos/farmacocinética , Ratas , Resultado del Tratamiento , Factor Trefoil-2
8.
Int Rev Immunol ; 28(6): 465-86, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19954359

RESUMEN

Lactic acid bacteria are a group of taxonomically diverse, Gram-positive food-grade bacteria that have been safely consumed throughout history. The lactic acid bacterium Lactococcus lactis, well-known for its use in the manufacture of cheese, can be genetically engineered and orally formulated to deliver therapeutic proteins in the gastrointestinal tract. This review focuses on the genetic engineering of Lactococcus lactis to secrete high-quality, correctly processed bioactive molecules derived from a eukaryotic background. The therapeutic applications of these genetically modified strains are discussed, with special regards to immunomodulation.


Asunto(s)
Tracto Gastrointestinal/inmunología , Ingeniería Genética/métodos , Lactococcus lactis/genética , Tejido Linfoide/inmunología , Animales , Evaluación Preclínica de Medicamentos , Tracto Gastrointestinal/microbiología , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Interleucina-10/genética , Interleucina-10/metabolismo , Lactococcus lactis/crecimiento & desarrollo , Lactococcus lactis/metabolismo , Péptidos/genética , Péptidos/metabolismo , Factor Trefoil-2
9.
J Immunol ; 183(4): 2390-6, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19635921

RESUMEN

Active delivery of recombinant autoantigens or allergens at the intestinal mucosa by genetically modified Lactococcus lactis (LL) provides a novel therapeutic approach for the induction of tolerance. Celiac disease is associated with either HLA-DQ2- or HLA-DQ8-restricted responses to specific antigenic epitopes of gliadin, and may be treated by induction of Ag-specific tolerance. We investigated whether oral administration of LL-delivered DQ8-specific gliadin epitope induces Ag-specific tolerance. LL was engineered to secrete a deamidated DQ8 gliadin epitope (LL-eDQ8d) and the induction of Ag-specific tolerance was studied in NOD AB degrees DQ8 transgenic mice. Tolerance was assessed by delayed-type hypersensitivity reaction, cytokine measurements, eDQ8d-specific proliferation, and regulatory T cell analysis. Oral administration of LL-eDQ8d induced suppression of local and systemic DQ8-restricted T cell responses in NOD AB degrees DQ8 transgenic mice. Treatment resulted in an Ag-specific decrease of the proliferative capacity of inguinal lymph node (ILN) cells and lamina propria cells. Production of IL-10 and TGF-beta and a significant induction of Foxp3(+) regulatory T cells were associated with the eDQ8d-specific suppression induced by LL-eDQ8d. These data provide support for the development of effective therapeutic approaches for gluten-sensitive disorders using orally administered Ag-secreting LL. Such treatments may be effective even in the setting of established hypersensitivity.


Asunto(s)
Epítopos de Linfocito T/inmunología , Gliadina/inmunología , Antígenos HLA-DQ/inmunología , Tolerancia Inmunológica , Epítopos Inmunodominantes/inmunología , Lactococcus lactis/inmunología , Fragmentos de Péptidos/inmunología , Administración Oral , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/terapia , Células Clonales , Modelos Animales de Enfermedad , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/genética , Gliadina/administración & dosificación , Gliadina/genética , Antígenos HLA-DQ/genética , Humanos , Tolerancia Inmunológica/genética , Inmunización , Epítopos Inmunodominantes/administración & dosificación , Epítopos Inmunodominantes/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Lactococcus lactis/genética , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Datos de Secuencia Molecular , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética
10.
Ann N Y Acad Sci ; 1182: 135-45, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20074282

RESUMEN

Interleukin-10 (IL-10) is central in immune downregulation, but so far its use in inflammatory diseases remains cumbersome. For treatment of inflammatory bowel disease, adequate amounts of IL-10 must reach the intestinal lining. Systemic injection of a pharmacologically active doses of recombinant human (rh) IL-10 results in very low mucosal levels of protein and severe toxicity and side effects. In animal models, topical and active delivery of IL-10 by ingestion of recombinant Lactococcus lactis (L. lactis) was shown to be a valuable alternative. Starting thereof we have developed a novel pharmaceutical platform. Our expertise and TopAct (topical and active) delivery technology allows use of recombinant L. lactis- ActoBiotics- in clinical practice. Here we discuss the development of recombinant L. lactis for intestinal delivery of rhIL-10 in humans.


Asunto(s)
Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Interleucina-10/administración & dosificación , Interleucina-10/metabolismo , Lactococcus lactis/genética , Animales , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Sistemas de Liberación de Medicamentos , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucina-10/genética , Mucosa Intestinal/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
11.
Inflamm Bowel Dis ; 14(4): 471-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18092343

RESUMEN

BACKGROUND: Genetically modified Lactococcus lactis secreting interleukin-10 (IL-10) has been demonstrated to provide localized delivery of a therapeutic agent through active in situ synthesis in murine colitis. At present, many aspects of the exact mechanism by which the beneficial effect of the IL-10-producing L. lactis on the mucosa is mediated remain to be clarified. METHODS: Our aim was to determine the interaction of L. lactis with the intestinal mucosa. Therefore, we administered IL-10-producing L. lactis to healthy mice and in 2 mouse models of chronic colitis. Paraffin sections of ileum and colon samples were examined with confocal and transmission electron microscopy. Ileum and colon homogenates were prepared after flushing and after removal of mucus layer and epithelium. These homogenates and homogenates of mesenteric lymph nodes and spleen were plated on agar and immunoblotting for L. lactis and IL-10 was performed. RESULTS: Both confocal and electron microscopy showed the presence of lactococci in inflamed intestinal mucosa of mice with colitis. We recovered viable bacteria that could still produce IL-10 from homogenates of inflamed ileum and colon of which mucous and epithelial layers were removed. We did not find lactococci in mesenteric lymph nodes or in the spleen of mice with colitis. CONCLUSIONS: This study demonstrates uptake of IL-10-secreting L. lactis by the paracellular route in inflamed mucosal tissue. We suggest that IL-10 production by L. lactis residing inside the mucosa in the vicinity of responsive cells can improve the local action of interleukin-10 in inflamed tissue and the efficiency of the treatment.


Asunto(s)
Colitis/patología , Interleucina-10/biosíntesis , Lactococcus lactis/metabolismo , Animales , Colitis/microbiología , Colitis/terapia , Colon/microbiología , Colon/patología , Ingeniería Genética , Íleon/microbiología , Íleon/patología , Interleucina-10/uso terapéutico , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Lactococcus lactis/genética , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Microscopía Electrónica de Transmisión
12.
Int J Exp Pathol ; 89(6): 438-46, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19134053

RESUMEN

Galectins are increasingly the focus of biomedical research. Although they are involved at different stages in inflammation, data on galectins in colitis remain scarce. The aim of this study was to determine and compare the expression of galectins in acute and chronic experimental colitis in mice. Immunohistochemistry for galectins-1, -3 and -4 was performed on colon tissue from C57BL/6 and BALB/c mice with acute dextran sodium sulphate colitis and from 129 Sv/Ev IL-10 knock-out (IL-10(-/-)) mice. From these three mouse strains, we first detected major differences in galectin expression related to the genetic background in the control animals. With regard to inflammation, chronic colitis in IL-10(-/-) mice was associated with increased galectin-4 expression; in contrast with the two other models, no galectin-1 and -3 alterations were observed in IL-10(-/-) mice. Acute colitis in C57BL/6 and BALB/c mice showed increased galectin-3 expression in the lamina propria and the crypt epithelium, together with a decreased nuclear expression. These results suggest an involvement of galectins in the development and perpetuation of colonic inflammation and illustrate that the choice of the mouse strain for studying galectins might influence the outcome of the experiments.


Asunto(s)
Colitis/metabolismo , Colon/química , Galectinas/análisis , Enfermedad Aguda , Animales , Enfermedad Crónica , Sulfato de Dextran , Femenino , Galectina 1/análisis , Galectina 3/análisis , Galectina 4/análisis , Inmunohistoquímica , Interleucina-10/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Especificidad de la Especie
13.
Gastroenterology ; 133(2): 517-28, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17681173

RESUMEN

BACKGROUND AND AIMS: Obtaining antigen-specific immune suppression is an important goal in developing treatments of autoimmune, inflammatory, and allergic gastrointestinal diseases. Oral tolerance is a powerful means for inducing tolerance to a particular antigen, but implementing this strategy in humans has been difficult. Active delivery of recombinant autoantigens or allergens at the intestinal mucosa by genetically modified Lactococcus lactis (L lactis) provides a novel therapeutic approach for inducing tolerance. METHODS: We engineered the food grade bacterium L lactis to secrete ovalbumin (OVA) and evaluated its ability to induce OVA-specific tolerance in OVA T-cell receptor (TCR) transgenic mice (DO11.10). Tolerance induction was assessed by analysis of delayed-type hypersensitivity responses, measurement of cytokines and OVA-specific proliferation, phenotypic analysis, and adoptive transfer experiments. RESULTS: Intragastric administration of OVA-secreting L lactis led to active delivery of OVA at the mucosa and suppression of local and systemic OVA-specific T-cell responses in DO11.10 mice. This suppression was mediated by induction of CD4(+)CD25(-) regulatory T cells that function through a transforming growth factor beta-dependent mechanism. Restimulation of splenocytes and gut-associated lymph node tissue from these mice resulted in a significant OVA-specific decrease in interferon gamma and a significant increase in interleukin-10 production. Furthermore, Foxp3 and CTLA-4 were significantly up-regulated in the CD4(+)CD25(-) population. CONCLUSIONS: Mucosal antigen delivery by oral administration of genetically engineered L lactis leads to antigen-specific tolerance. This approach can be used to develop effective therapeutics for systemic and intestinal immune-mediated inflammatory diseases.


Asunto(s)
Hipersensibilidad Tardía/inmunología , Tolerancia Inmunológica , Intestinos/inmunología , Lactococcus lactis/metabolismo , Ovalbúmina/inmunología , Probióticos/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Administración Oral , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , Antígeno CTLA-4 , Proliferación Celular , Relación Dosis-Respuesta Inmunológica , Femenino , Factores de Transcripción Forkhead/metabolismo , Hipersensibilidad Tardía/metabolismo , Inmunidad Mucosa , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/análisis , Mucosa Intestinal/metabolismo , Intestinos/citología , Lactococcus lactis/genética , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/biosíntesis , Ovalbúmina/genética , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Probióticos/administración & dosificación , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta/metabolismo
14.
J Immunol ; 179(4): 2300-9, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17675491

RESUMEN

The glycosphingolipid alpha-galactosylceramide (alpha-GalCer) has been shown to be a potent activator of invariant NKT (iNKT) cells, rapidly inducing large amounts of both Th1 and Th2 cytokines upon injection in mice. The C-glycoside analog of alpha-GalCer (alpha-C-GalCer), by contrast, results in an enhanced Th1-type response upon activation of iNKT cells. We administered a single dose of these Ags to DBA/1 mice during the early induction phase of collagen-induced arthritis and demonstrated therapeutic efficacy of alpha-GalCer when administered early rather than late during the disease. Surprisingly, the Th1-polarizing analog alpha-C-GalCer also conferred protection. Furthermore, a biphasic role of IFN-gamma in the effect of iNKT cell stimulation was observed. Whereas in vivo neutralization of IFN-gamma release induced by either alpha-GalCer or alpha-C-GalCer early during the course of disease resulted in partial improvement of clinical arthritis symptoms, blockade of IFN-gamma release later on resulted in a more rapid onset of arthritis. Although no phenotypic changes in conventional T cells, macrophages, or APCs could be detected, important functional differences in T cell cytokine production in serum were observed upon polyclonal T cell activation, 2 wk after onset of arthritis. Whereas alpha-GalCer-treated mice produced significantly higher amounts of IL-10 upon systemic anti-CD3 stimulation compared with PBS controls, T cells from alpha-C-GalCer-treated mice, by contrast, produced substantially lower levels of cytokines, suggesting the involvement of different protective mechanisms. In conclusion, these findings suggest long-term, ligand-specific, time-dependent, and partially IFN-gamma-dependent immunomodulatory effects of iNKT cells in collagen-induced arthritis.


Asunto(s)
Artritis Experimental/prevención & control , Galactosilceramidas/administración & dosificación , Factores Inmunológicos/administración & dosificación , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Células TH1/inmunología , Células Th2/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/patología , Artritis Experimental/inmunología , Artritis Experimental/patología , Complejo CD3/inmunología , Interferón gamma/inmunología , Interleucina-10/inmunología , Células Asesinas Naturales/patología , Ligandos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos DBA , Células TH1/patología , Células Th2/patología , Factores de Tiempo
15.
Appl Environ Microbiol ; 72(12): 7694-700, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17028239

RESUMEN

Interleukin-10 (IL-10) is a promising candidate for the treatment of inflammatory bowel disease. Intragastric administration of Lactococcus lactis genetically modified to secrete IL-10 in situ in the intestine was shown to be effective in healing and preventing chronic colitis in mice. However, its use in humans is hindered by the sensitivity of L. lactis to freeze-drying and its poor survival in the gastrointestinal tract. We expressed the trehalose synthesizing genes from Escherichia coli under control of the nisin-inducible promoter in L. lactis. Induced cells accumulated intracellular trehalose and retained nearly 100% viability after freeze-drying, together with a markedly prolonged shelf life. Remarkably, cells producing trehalose were resistant to bile, and their viability in human gastric juice was enhanced. None of these effects were seen with exogenously added trehalose. Trehalose accumulation did not interfere with IL-10 secretion or with therapeutic efficacy in murine colitis. The newly acquired properties should enable a larger proportion of the administered bacteria to reach the gastrointestinal tract in a bioactive form, providing a means for more effective mucosal delivery of therapeutics.


Asunto(s)
Colitis/terapia , Tracto Gastrointestinal/microbiología , Lactococcus lactis/crecimiento & desarrollo , Trehalosa/metabolismo , Animales , Bilis/química , Enfermedad Crónica , Colitis/microbiología , Crioprotectores/metabolismo , Crioprotectores/farmacología , Sistemas de Liberación de Medicamentos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Femenino , Liofilización , Jugo Gástrico/química , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Lactococcus lactis/efectos de los fármacos , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trehalosa/farmacología
16.
Ann N Y Acad Sci ; 1072: 176-86, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17057198

RESUMEN

Food-grade bacteria have been consumed throughout history without associated pathologies and are, therefore, absolutely safe to ingest. Unexpectedly, Lactococcus lactis (L. lactis), known from cheese production, can be genetically engineered to constantly secrete satisfactory amounts of bioactive cytokines. Both of these features enabled the development of a new kind of topical delivery system: topical and active delivery of therapeutic proteins by genetically modified micro-organisms. The host organism's record inspired the development of applications that target intestinal diseases. In a variety of mouse models, chronic colon inflammation can be successfully treated with (interleukin) IL-10-secreting L. lactis. Trefoil factor (TFF) producer strains have also been shown to be very effective in the treatment of acute colitis. Such novel therapeutic strains are textbook examples of genetically modified (GM) organisms. There are legitimate concerns with regard to the deliberate release of GM micro-organisms. On development of these applications, therefore, we have engineered these bacteria in such a way that biological containment is guaranteed. The essential gene thyA, encoding thymidylate synthase, has been exchanged for IL-10. This makes the GM strain critically dependent on thymidine. Lack of thymidine, for example, resulting from thymidine consumption by thyA-deficient strains-will irreversibly lead to induced "thymidine-less death." This accomplishment has created the possibility of using this strategy for application in human medicine.


Asunto(s)
Lactococcus lactis/genética , Animales , Anticuerpos/uso terapéutico , Citocinas/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Humanos , Modelos Animales
17.
Arthritis Rheum ; 54(6): 1856-66, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16736523

RESUMEN

OBJECTIVE: The advent of tumor necrosis factor (TNF)-blocking drugs has provided rheumatologists with an effective, but highly expensive, treatment for the management of established rheumatoid arthritis (RA). Our aim was to explore preclinically the application of camelid anti-TNF VHH proteins, which are single-domain antigen binding (VHH) proteins homologous to human immunoglobulin V(H) domains, as TNF antagonists in a mouse model of RA. METHODS: Llamas were immunized with human and mouse TNF, and antagonistic anti-TNF VHH proteins were isolated and cloned for bacterial production. The resulting anti-TNF VHH proteins were recombinantly linked to yield bivalent mouse and human TNF-specific molecules. To increase the serum half-life and targeting properties, an anti-serum albumin anti-TNF VHH domain was incorporated into the bivalent molecules. The TNF-neutralizing potential was analyzed in vitro. Mouse TNF-specific molecules were tested in a therapeutic protocol in murine collagen-induced arthritis (CIA). Disease progression was evaluated by clinical scoring and histologic evaluation. Targeting properties were evaluated by 99mTc labeling and gamma camera imaging. RESULTS: The bivalent molecules were up to 500 times more potent than the monovalent molecules. The antagonistic potency of the anti-human TNF VHH proteins exceeded even that of the anti-TNF antibodies infliximab and adalimumab that are used clinically in RA. Incorporation of binding affinity for albumin into the anti-TNF VHH protein significantly prolonged its serum half-life and promoted its targeting to inflamed joints in the murine CIA model of RA. This might explain the excellent therapeutic efficacy observed in vivo. CONCLUSION: These data suggest that because of the flexibility of their format, camelid anti-TNF VHH proteins can be converted into potent therapeutic agents that can be produced and purified cost-effectively.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis Experimental/terapia , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Pesadas de Inmunoglobulina/uso terapéutico , Región Variable de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/uso terapéutico , Factor de Necrosis Tumoral alfa/inmunología , Adalimumab , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Camélidos del Nuevo Mundo/inmunología , Semivida , Cadenas Pesadas de Inmunoglobulina/sangre , Región Variable de Inmunoglobulina/sangre , Infliximab , Ratones , Ratones Endogámicos BALB C
18.
Clin Gastroenterol Hepatol ; 4(6): 754-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16716759

RESUMEN

BACKGROUND & AIMS: The use of living, genetically modified bacteria is an effective approach for topical delivery of immunomodulatory proteins. This strategy circumvents systemic side effects and allows long-term treatment of chronic diseases. However, treatment of patients with a living, genetically modified bacterium raises questions about the safety for human subjects per se and the biologic containment of the transgene. METHODS: We treated Crohn's disease patients with genetically modified Lactococcus lactis (LL-Thy12) in which the thymidylate synthase gene was replaced with a synthetic sequence encoding mature human interleukin-10. Ten patients were included in a placebo-uncontrolled trial. Patients were assessed daily for the presence of potential adverse effects by direct questioning and assessment of disease activity. We evaluated the presence and kinetics of LL-Thy12 release in the stool of patients by conventional culturing and quantitative polymerase chain reaction of LL-Thy12 gene sequences. RESULTS: Treatment with LL-Thy12 was safe because only minor adverse events were present, and a decrease in disease activity was observed. Moreover, fecally recovered LL-Thy12 bacteria were dependent on thymidine for growth and interleukin-10 production, indicating that the containment strategy was effective. CONCLUSIONS: Here we show that the use of genetically modified bacteria for mucosal delivery of proteins is a feasible strategy in human beings. This novel strategy avoids systemic side effects and is biologically contained; therefore it is suitable as maintenance treatment for chronic intestinal disease.


Asunto(s)
Enfermedad de Crohn/terapia , Terapia Genética , Interleucina-10/administración & dosificación , Lactococcus lactis , Proteína C-Reactiva/análisis , Enfermedad de Crohn/patología , Humanos , Interleucina-10/genética , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Organismos Modificados Genéticamente , Comprimidos Recubiertos , Timidilato Sintasa/genética , Transgenes
19.
Free Radic Biol Med ; 40(2): 285-94, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16413410

RESUMEN

Using tumor cell-restricted overexpression of glutathione peroxidase 4 (GP x 4), we investigated the contribution of tumor cell eicosanoids to solid tumor growth and malignant progression in two tumor models differing in tumorigenic potential. By lowering cellular lipid hydroperoxide levels, GP x 4 inhibits cyclooxygenase (COX) and lipoxygenase (LOX) activities. GP x 4 overexpression drastically impeded solid tumor growth of weakly tumorigenic L929 fibrosarcoma cells, whereas B16BL6 melanoma solid tumor growth was unaffected. Yet, GP x 4 overexpression did markedly increase the sensitivity of B16BL6 tumors to angio-destructive TNF-alpha therapy and abolished the metastatic lung colonizing capacity of B16BL6 cells. Furthermore, the GP x 4-mediated suppression of tumor cell prostaglandin E(2) (PGE(2)) production impeded the induction of COX-2 expression by the tumor stress conditions hypoxia and inflammation. Thus, our results reflect a PGE(2)-driven positive feedback loop for COX-2 expression in tumor cells. This was further supported by the restoration of COX-2 induction capacity of GP x 4-overexpressing L929 tumor cells when cultured in the presence of exogenous PGE(2). Thus, although COX-2 expression and eicosanoid production may be enabled by PGE(2) from the tumor microenvironment, our results demonstrate the predominant tumor cell origin of protumoral eicosanoids, promoting solid tumor growth of weakly tumorigenic tumors and malignant progression of strongly tumorigenic tumors.


Asunto(s)
Eicosanoides/biosíntesis , Fibrosarcoma/fisiopatología , Glutatión Peroxidasa/metabolismo , Melanoma/fisiopatología , Metástasis de la Neoplasia/prevención & control , Animales , Hipoxia de la Célula/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/antagonistas & inhibidores , Dinoprostona/biosíntesis , Modelos Animales de Enfermedad , Eicosanoides/antagonistas & inhibidores , Femenino , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Transferencia de Gen , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/farmacología , Lipooxigenasa/efectos de los fármacos , Lipooxigenasa/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Porcinos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/uso terapéutico
20.
Eur J Pharm Biopharm ; 61(3): 134-41, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15967652

RESUMEN

Layering of recombinant hIL-10 producing Lactococcus lactis (L. lactis Thy12) on inert carriers is a promising technique for the preparation of a multi-particulate formulation of viable, hIL-10 producing L. lactis. To improve viability after layering and storage, L. lactis Thy12 was layered in different matrices (10% skim milk and/or 2.5, 5, 10% inulin). After layering, the highest viability was obtained in the 10% skim milk supplemented with 5% inulin matrix (8.7%). However, upon storage, 10% skim milk alone yielded the highest viability. Thereby, layered L. lactis Thy12 showed superior long term stability in comparison with freeze-dried L. lactis Thy12. The layering process was performed during 3h without encountering technical problems, with good layer consistence and constant viability. Enteric properties were obtained with a 30% Eudragit L30D-55 or 15% Eudragit FS30D coating and maintained during an initial six months storage period (-20 degrees C/20% RH). After in vitro simulation of the gastric stage, only 5% of the bacteria remained viable in Eudragit L30D-55 coated pellets, contrary to 85% in Eudragit FS30D coated pellets, indicating its superior protective capacity against gastric fluid. After eight months storage (-20 degrees C), 80% of the initial L. lactis Thy12 remained viable in the Eudragit FS30D coated pellets.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Íleon/metabolismo , Interleucina-10/administración & dosificación , Lactococcus lactis/genética , Química Farmacéutica , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Interleucina-10/biosíntesis , Lactococcus lactis/fisiología , Ácidos Polimetacrílicos/administración & dosificación , Comprimidos Recubiertos , Tecnología Farmacéutica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...