Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunology ; 170(4): 540-552, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37671510

RESUMEN

Adoptive regulatory T-cell (Treg) transfer has emerged as a promising therapeutic strategy for regulating immune responses in organ transplantation, graft versus host disease, and autoimmunity, including Type 1 diabetes. Traditionally, Treg for adoptive therapy have been sorted and expanded in vitro using high doses of IL-2, demonstrating stability and suppressive capabilities. However, limitations in their long-term survival post-infusion into patients have been observed. To address this challenge, we investigated a novel expansion protocol incorporating interleukin-7 (IL-7) alongside the traditional method utilizing IL-2 (referred to as IL-7 method, IL-7M). Our study revealed that naïve Treg express significant levels of CD127 and display robust responsiveness to IL-7, characterized by STAT-5 phosphorylation. Expanding naïve Treg with the IL-7M protocol led to a substantial enrichment of CD45RA+ CD62L+ CD95+ Treg but showing a reduction in the final cell yield and suppressive function. Moreover, Treg expanded with the IL-7M exhibited preserved telomere length and demonstrated enhanced resistance to cytokine withdrawal and fas-mediated apoptosis. When transferred into NSG mice IL-7M-Treg persisted longer and reduced the expansion of T cells, but did not significantly reduce the severity of xenoGvHD. In conclusion, our data demonstrate the feasibility of expanding naïve Treg in the presence of IL-7 to generate a Treg product enriched in poorly differentiated CD45RA+ cells with enhanced survival capabilities.


Asunto(s)
Interleucina-7 , Linfocitos T Reguladores , Humanos , Ratones , Animales , Interleucina-2 , Citocinas , Traslado Adoptivo/métodos , Antígenos Comunes de Leucocito , Factores de Transcripción Forkhead
2.
Opt Lett ; 48(12): 3183-3186, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37319057

RESUMEN

In this Letter, we present a scheme for detecting fiber-bending eavesdropping based on feature extraction and machine learning (ML). First, 5-dimensional features from the time-domain signal are extracted from the optical signal, and then a long short-term memory (LSTM) network is applied for eavesdropping and normal event classification. Experimental data are collected from a 60 km single-mode fiber transmission link with eavesdropping implemented by a clip-on coupler. Results show that the proposed scheme achieves a 95.83% detection accuracy. Furthermore, since the scheme focuses on the time-domain waveform of the received optical signal, additional devices and a special link design are not required.


Asunto(s)
Aprendizaje Automático , Redes Neurales de la Computación
3.
Immunology ; 167(3): 303-313, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35752961

RESUMEN

Autoreactive T cells with the phenotype and function of different memory subsets are present in patients who developed type 1 diabetes (TID). According to the progressive differentiation model, memory subsets generate from naïve precursors in a linear and unidirectional path depending on the strength and quality of stimulatory signals. By observing human naïve T cells in contact with GAD65 loaded autologous dendritic cells, we observed that approximately 10% of cells divided with the plane of cell division parallel to the one of the immune synapse, causing phenotypic asymmetries in the proximal and distal daughter T cells. After the first T cell division, proximal and distal daughter T cells showed different phenotype, metabolic signature and commitment to differentiate towards long-lived memory T cells or T cells with effector function. Subjects with or without T1D showed a similar frequency of asymmetric T cell division (ATCD) for autoantigens and recall antigens specific T cells, however the frequency of ATCD is significantly increased in autoreactive T cells in patients with T1D when IL-7 was added to the culture. An increased upregulation of GLUT1 in response to IL-7 in patients with T1D was related to the rate of ATCD. Our results showed that ATCD is associated with an early divergence in the differentiation fate of naïve T cells specific for GAD65 during first antigen encounter.


Asunto(s)
Diabetes Mellitus Tipo 1 , Autoantígenos , Diferenciación Celular , División Celular , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Memoria Inmunológica , Interleucina-7/metabolismo , Células T de Memoria , Subgrupos de Linfocitos T
4.
Int J Mol Sci ; 20(19)2019 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-31597342

RESUMEN

An increasing body of evidence indicates that bio-energetic metabolism of T cells can be manipulated to control T cell responses. This potentially finds a field of application in the control of the T cell responses in autoimmune diseases, including in type 1 diabetes (T1D). Of the possible metabolic targets, Glut1 gained considerable interest because of its pivotal role in glucose uptake to fuel glycolysis in activated T cells, and the recent development of a novel class of small molecules that act as selective inhibitor of Glut1. We believe we can foresee a possible application of pharmacological Glut1 blockade approach to control autoreactive T cells that destroy insulin producing beta cells. However, Glut1 is expressed in a broad range of cells in the body and off-target and side effect are possible complications. Moreover, the duration of the treatment and the age of patients are critical aspects that need to be addressed to reduce toxicity. In this paper, we will review recent literature to determine whether it is possible to design a pharmacological Glut1 blocking strategy and how to apply this to autoimmunity in T1D.


Asunto(s)
Autoinmunidad , Transportador de Glucosa de Tipo 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/química , Transportador de Glucosa de Tipo 1/genética , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Terapia Molecular Dirigida , Transducción de Señal , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos
5.
Curr Diab Rep ; 17(5): 24, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28303386

RESUMEN

PURPOSE OF REVIEW: An increasing body of evidence indicates that bio-energetic metabolism of activated T cells is a potential target to control the autoimmune response in type 1 diabetes (T1D). RECENT FINDINGS: T-cell activation and proliferation is linked to the cell capacity to provide sufficient energy and biosynthesis molecules to support T-cell growth and division. This makes T cells susceptible to metabolic inhibition for the control of the T-cell response. There is a wide therapeutic arsenal of metabolic inhibitors, including novel classes of drugs that have become recently available. With the current knowledge and availability of metabolic inhibitors, we are now in the position to design a metabolic inhibition strategy to determine whether targeting of autoreactive T cells is an effective strategy to control the process of ß-cell destruction in T1D.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Células Secretoras de Insulina/inmunología , Linfocitos T/metabolismo , Animales , Diabetes Mellitus Tipo 1/inmunología , Humanos , Activación de Linfocitos , Redes y Vías Metabólicas , Linfocitos T/inmunología
7.
Curr Diab Rep ; 16(5): 40, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26983628

RESUMEN

Immunomodulation of the autoreactive T cell response is considered a major strategy to control beta-cell autoimmunity, both in the natural history of type 1 diabetes and in islet transplantation, which can be affected by autoimmunity recurrence. So far, these strategies have had modest results, prompting efforts to define novel cellular and molecular targets to control autoreactive T cell expansion and activation. Novel findings highlighted the important role of the homeostatic cytokine interleukin-7 in inducing proliferation and differentiation of autoreactive T cell clones that causes beta-cell autoimmunity. In this review, we discuss recent evidences and novel findings on the role of IL-7 mediated homeostatic T cell proliferation in the process of beta-cell destruction and evidences of how targeting IL-7 and its receptor could be an innovative and effective strategy to control beta-cell autoimmunity.


Asunto(s)
Autoinmunidad , Proliferación Celular , Homeostasis , Células Secretoras de Insulina/inmunología , Linfocitos T/inmunología , Animales , Humanos , Interleucina-7/inmunología , Linfocitos T/citología
8.
Curr Diab Rep ; 14(9): 518, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24996523

RESUMEN

Antigen-experienced T-cells directly target and destroy insulin-producing beta cells in patients with Type 1 diabetes. Consequently, T-cells are also major targets of immunomodulatory strategies that aim to prevent or delay the immune mediated loss of islet beta-cell function. These strategies have had modest success, prompting efforts into better defining the mechanisms that drive the differentiation of quiescent autoreactive clones into pathogenic effector and memory T-cells. Recent and novel findings now indicate that in addition to the classic mechanisms of antigenic recognition, autoreactive T-cell differentiation and expansion can be boosted by the homeostatic cytokine interleukin-7. In this article, we discuss recent evidence of the role of IL-7 mediated T-cell proliferation in the pathogenesis of Type 1 diabetes and the rationale for including immunomodulatory molecules targeting the IL-7/IL-7R axis in immunotherapeutic strategies to control beta-cell autoimmunity.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Interleucina-7/metabolismo , Animales , Autoinmunidad/inmunología , Humanos , Activación de Linfocitos/inmunología , Receptores de Interleucina-7/metabolismo , Linfocitos T/inmunología
9.
J Immunol ; 189(12): 5649-58, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23129754

RESUMEN

CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) control the activation and expansion of alloreactive and autoreactive T cell clones. Because uncontrolled activation and expansion of autoreactive T cells occur in an IL-7-rich environment, we explored the possibility that IL-7 may affect the function of Treg. We show that the functional high-affinity IL-7R is expressed on both naive and memory Tregs, and exposure to IL-7 results in STAT-5 phosphorylation. Naive, but not memory, Tregs proliferated greatly and acquired a memory phenotype in the setting of a suppression assay when IL-7 was present. Importantly, the presence of IL-7 abrogated the capacity of Tregs to suppress proliferation of conventional T cells in response to TCR activators, including alloantigens and autoantigens. Removal of IL-7 restored the suppressive function of Tregs. Preblocking of the IL-7R on the Tregs also restored suppressor function, indicating that IL-7 directly affected Treg function. Thus, prolonged periods of homeostatic expansion can temporarily release natural regulatory brakes on T cells, thereby providing an additional mechanism for activating and expanding alloreactive and autoreactive T cells.


Asunto(s)
Autoantígenos/biosíntesis , Antígenos CD4/biosíntesis , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/biosíntesis , Interleucina-7/fisiología , Isoantígenos/biosíntesis , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Proliferación Celular , Células Cultivadas , Contraindicaciones , Humanos , Inmunofenotipificación , Interleucina-10/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo
10.
Environ Pollut ; 358: 124471, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38950846

RESUMEN

Associations between indoor air pollution from fine particulate matter (PM with aerodynamic diameter dp < 2.5 µm) and human health are poorly understood. Here, we analyse the concentration-response curves for fine and ultrafine PM, the gene expression, and the methylation patterns in human bronchial epithelial cells (BEAS-2B) exposed at the air-liquid interface (ALI) within a classroom in downtown Rome. Our results document the upregulation of aryl hydrocarbon receptor (AhR) and genes associated with xenobiotic metabolism (CYP1A1 and CYP1B1) in response to single exposure of cells to fresh urban aerosols at low fine PM mass concentrations within the classroom. This is evidenced by concentrations of ultrafine particles (UFPs, dp < 0.1 µm), polycyclic aromatic hydrocarbons (PAH), and ratios of black carbon (BC) to organic aerosol (OA). Additionally, an interleukin 18 (IL-18) down-regulation was found during periods of high human occupancy. Despite the observed gene expression dysregulation, no changes were detected in the methylation levels of the promoter regions of these genes, indicating that the altered gene expression is not linked to changes in DNA methylation and suggesting the involvement of another epigenetic mechanism in the gene regulation. Gene expression changes at low exposure doses have been previously reported. Here, we add the possibility that lung epithelial cells, when singly exposed to real environmental concentrations of fine PM that translate into ultra-low doses of treatment, may undergo epigenetic alteration in the expression of genes related to xenobiotic metabolism. Our findings provide a perspective for future indoor air quality regulations. We underscore the potential role of indoor UFPs as carriers of toxic molecules with low-pressure weather conditions, when rainfall and strong winds may favour low levels of fine PM.

11.
Clin Dev Immunol ; 2013: 217934, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23970924

RESUMEN

Pancreatic islet transplantation in patients with type 1 diabetes mellitus is performed under immunosuppression to avoid alloreactive T cell responses and to control the reactivation of autoreactive memory T cells. However, lymphopenia associated with immunosuppression and T cell depletion can induce a paradoxical expansion of lymphocyte subsets under the influence of homeostatic proliferation. Homeostatic T cell proliferation is mainly driven by the IL-7/IL-7 receptor axis, a molecular pathway which is not affected by standard immune-suppressive drugs and, consequently, represents a novel potential target for immuno-modulatory strategies. In this review, we will discuss how homeostatic T cell proliferation can support autoimmunity recurrence after islet transplantation and how it can be targeted by new therapeutic approaches.


Asunto(s)
Homeostasis , Trasplante de Islotes Pancreáticos/inmunología , Activación de Linfocitos , Subgrupos de Linfocitos T/inmunología , Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Humanos , Memoria Inmunológica , Terapia de Inmunosupresión , Interleucina-7/metabolismo , Recurrencia
12.
Exp Fluids ; 64(4): 80, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37016621

RESUMEN

Due to its importance in airborne disease transmission, especially because of the COVID-19 pandemic, much attention has recently been devoted by the scientific community to the analysis of dispersion of particle-laden air clouds ejected by humans during different respiratory activities. In spite of that, a lack of knowledge is still present particularly with regard to the velocity of the emitted particles, which could differ considerably from that of the air phase. The velocity of the particles is also expected to vary with their size. In this work, simultaneous measurements of size and velocity of particles emitted by humans while speaking have been performed by means of Interferometric Laser Imaging Droplet Sizing (ILIDS). This technique allowed us to detect emitted particles with size down to 2 µm as well as to quantify all three components of the velocity vector and the particle concentration. The outcomes of this work may be used as boundary conditions for numerical simulations of infected respiratory cloud transmission.

13.
Front Endocrinol (Lausanne) ; 14: 1175640, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37409229

RESUMEN

Aim: In a recent randomized, multicenter trial (NCT02814838) a short-term anti-inflammatory treatment with ladarixin (LDX; an inhibitor of the CXCR1/2 chemokine receptors) did not show benefit on preserving residual beta cell function in new-onset type 1 diabetes. We present a post hoc analysis of trial patients in the predefined subgroup analysis developed according to baseline daily insulin requirement (DIR) tertiles. Method: A double-blind, randomized (2:1), placebo-controlled study was conducted in 45 men and 31 women (aged 18-46 years) within 100 days of the first insulin administration. Patients received LDX (400 mg twice daily) for three cycles of 14 days on/14 days off, or placebo. The primary endpoint was the area under the curve for C-peptide [AUC (0-120 min)] in response to a 2-h mixed meal tolerance test (MMTT) at week 13 ± 1. Seventy-five patients completed the week 13 MMTT and were divided into three groups according to the DIR tertiles: lower, ≤ 0.23U/kg/die (n = 25); middle, 0.24-0.40 U/kg/die (n = 24); upper, ≥ 0.41 U/kg/die (n = 26). Results: When considering the patients in the upper tertile (HIGH-DIR), C-peptide AUC (0-120 min) at 13 weeks was higher in the LDX group (n = 16) than in the placebo (n = 10) group [difference: 0.72 nmol/L (95% CI 0.9-1.34), p = 0.027]. This difference reduced over time (0.71 nmol/L at 26 weeks, p = 0.04; 0.42 nmol/L at 52 weeks, p = 0.29), while it has never been significant at any time in patients in the lower and/or middle tertile (LOW-DIR). We characterized at baseline the HIGH-DIR and found that endo-metabolic (HOMA-B, adiponectin, and glucagon-to-C-peptide ratio) and immunologic (chemokine (C-C motif) ligand 2 (CCL2)/monocyte chemoattractant protein 1 (MCP1) and Vascular Endothelial Growth Factor (VEGF)) features distinguished this group from LOW-DIR. Conclusion: While LDX did not prevent the progressive loss of beta-cell function in the majority of treated subjects, the post hoc analysis suggests that it could work in subjects with HIGH-DIR at baseline. As we found differences in endo-metabolic and immunologic parameters within this subgroup, this generates the hypothesis that the interactions between host factors and drug action can contribute to its efficacy. Further research is needed to evaluate this hypothesis.


Asunto(s)
Diabetes Mellitus Tipo 1 , Masculino , Humanos , Femenino , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Péptido C/metabolismo , Estudios Prospectivos , Factor A de Crecimiento Endotelial Vascular , Insulina/uso terapéutico
14.
Front Immunol ; 14: 1323439, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38077372

RESUMEN

Type 1 diabetes (T1D) presents a persistent medical challenge, demanding innovative strategies for sustained glycemic control and enhanced patient well-being. Beta cells are specialized cells in the pancreas that produce insulin, a hormone that regulates blood sugar levels. When beta cells are damaged or destroyed, insulin production decreases, which leads to T1D. Allo Beta Cell Transplantation has emerged as a promising therapeutic avenue, with the goal of reinstating glucose regulation and insulin production in T1D patients. However, the path to success in this approach is fraught with complex immunological hurdles that demand rigorous exploration and resolution for enduring therapeutic efficacy. This exploration focuses on the distinct immunological characteristics inherent to Allo Beta Cell Transplantation. An understanding of these unique challenges is pivotal for the development of effective therapeutic interventions. The critical role of glucose regulation and insulin in immune activation is emphasized, with an emphasis on the intricate interplay between beta cells and immune cells. The transplantation site, particularly the liver, is examined in depth, highlighting its relevance in the context of complex immunological issues. Scrutiny extends to recipient and donor matching, including the utilization of multiple islet donors, while also considering the potential risk of autoimmune recurrence. Moreover, unanswered questions and persistent gaps in knowledge within the field are identified. These include the absence of robust evidence supporting immunosuppression treatments, the need for reliable methods to assess rejection and treatment protocols, the lack of validated biomarkers for monitoring beta cell loss, and the imperative need for improved beta cell imaging techniques. In addition, attention is drawn to emerging directions and transformative strategies in the field. This encompasses alternative immunosuppressive regimens and calcineurin-free immunoprotocols, as well as a reevaluation of induction therapy and recipient preconditioning methods. Innovative approaches targeting autoimmune recurrence, such as CAR Tregs and TCR Tregs, are explored, along with the potential of stem stealth cells, tissue engineering, and encapsulation to overcome the risk of graft rejection. In summary, this review provides a comprehensive overview of the inherent immunological obstacles associated with Allo Beta Cell Transplantation. It offers valuable insights into emerging strategies and directions that hold great promise for advancing the field and ultimately improving outcomes for individuals living with diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Insulinas , Trasplante de Islotes Pancreáticos , Humanos , Células Secretoras de Insulina/metabolismo , Glucosa
16.
Sci Total Environ ; 807(Pt 2): 150490, 2022 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-34666221

RESUMEN

Air pollution and heat stress are major concerns associated with the liveability, resilience and sustainability of cities. They directly affect health and comfort and are associated with augmented morbidity and mortality and an increase in the energy demand for building ventilation, air cleaning and cooling. Nevertheless, the detrimental effects of poor air quality may partly be mitigated by increased urban ventilation. This strategy is closely related to the level of urbanization and the urban morphology. Therefore, detailed investigations on the impact of different morphologies on urban ventilation are of paramount importance. Computational Fluid Dynamics simulations have been widely used during the last decades to investigate the effects of the urban morphology on the urban ventilation. However, most of these studies focused on idealized building arrangements, while detailed investigations about the role of real urban morphologies are scarce. This study investigates the ventilation in a compact area in the city of Rome, Italy. 3D steady-state Reynolds-averaged Navier-Stokes simulations are performed to analyze the impact of Morphological Parameters (MP) on the urban ventilation. The results show a considerable worsening of urban ventilation with increasing building density with a reduction in the mean wind velocity up to 62% experienced at the pedestrian level (zp). Correlations between five MPs, e.g., plan area density, area-weighted mean building height, volume density, façade area density, and non-dimensional mean velocity at pedestrian level and at 10 m height are evaluated, and simple models are obtained using linear regression analysis. Among the selected MPs, the building façade area density shows a remarkable correlation with the non-dimensional mean velocity at zp (R2 = 0.82). Such correlations can be valuable tools for practitioners and urban designers, particularly during the first stage of planning, for highlighting areas potentially vulnerable to poor air conditions without running computationally expensive simulations.


Asunto(s)
Contaminación del Aire , Respiración , Ciudades , Ciudad de Roma , Ventilación
17.
Cell Rep ; 40(13): 111423, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36170817

RESUMEN

Induced pluripotent stem cells (iPSCs) represent a source from which ß cells can be derived for diabetes replacement therapy. However, their application may be hindered by immune-mediated responses. Although abrogation of major histocompatibility complex class I (MHC-I) can address this issue, it may trigger natural killer (NK) cells through missing-self recognition mechanisms. By profiling the relevant NK-activating ligands on iPSCs during in vitro differentiation into pancreatic ß cells, we find that they express high levels of B7-H3 and CD155. Hypothesizing that such surface ligands could be involved in the amplification of NK-activating signals following missing-self, we generate MHC-I-deprived B7-H3-/-, CD155-/-, and B7-H3-/-/CD155-/- iPSCs. All engineered lines correctly differentiate into insulin-secreting ß cells and are protected from cell lysis mediated by CD16dim and CD16+ NK subpopulations both in vitro and in vivo in NSG mice. Our data support targeted disruption of NK-activating ligands to enhance the transplant compatibility of MHC-I-/- iPSC pancreatic derivatives.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Secretoras de Insulina , Insulinas , Animales , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/metabolismo , Ligandos , Ratones
18.
J Clin Invest ; 118(5): 1806-14, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18431516

RESUMEN

Successful transplantation requires the prevention of allograft rejection and, in the case of transplantation to treat autoimmune disease, the suppression of autoimmune responses. The standard immunosuppressive treatment regimen given to patients with autoimmune type 1 diabetes who have received an islet transplant results in the loss of T cells. In many other situations, the immune system responds to T cell loss through cytokine-dependant homeostatic proliferation of any remaining T cells. Here we show that T cell loss after islet transplantation in patients with autoimmune type 1 diabetes was associated with both increased serum concentrations of IL-7 and IL-15 and in vivo proliferation of memory CD45RO(+) T cells, highly enriched in autoreactive glutamic acid decarboxylase 65-specific T cell clones. Immunosuppression with FK506 and rapamycin after transplantation resulted in a chronic homeostatic expansion of T cells, which acquired effector function after immunosuppression was removed. In contrast, the cytostatic drug mycophenolate mofetil efficiently blocked homeostatic T cell expansion. We propose that the increased production of cytokines that induce homeostatic expansion could contribute to recurrent autoimmunity in transplanted patients with autoimmune disease and that therapy that prevents the expansion of autoreactive T cells will improve the outcome of islet transplantation.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Interleucina-15/inmunología , Interleucina-7/inmunología , Trasplante de Islotes Pancreáticos , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Adulto , Animales , Diabetes Mellitus Tipo 1/sangre , Homeostasis , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Interleucina-15/sangre , Interleucina-7/sangre , Masculino , Ratones , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Ácido Micofenólico/uso terapéutico , Sirolimus/farmacología , Sirolimus/uso terapéutico , Linfocitos T/efectos de los fármacos , Tacrolimus/farmacología , Tacrolimus/uso terapéutico
19.
Diabetes ; 70(1): 171-181, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33122392

RESUMEN

The induction of antigen (Ag)-specific tolerance represents a therapeutic option for autoimmune diabetes. We demonstrated that administration of a lentiviral vector enabling expression of insulin B chain 9-23 (InsB9-23) (LV.InsB) in hepatocytes arrests ß-cell destruction in prediabetic NOD mice by generating InsB9-23-specific FoxP3+ T regulatory cells (Tregs). LV.InsB in combination with a suboptimal dose of anti-CD3 monoclonal antibody (combined therapy [CT], 1 × 5 µg [CT5]) reverts diabetes and prevents recurrence of autoimmunity after islet transplantation in ∼50% of NOD mice. We investigated whether CT optimization could lead to abrogation of recurrence of autoimmunity. Therefore, alloislets were transplanted after optimized CT tolerogenic conditioning (1 × 25 µg [CT25]). Diabetic NOD mice conditioned with CT25 when glycemia was <500 mg/dL remained normoglycemic for 100 days after alloislet transplantation and displayed reduced insulitis, but independently from the graft. Accordingly, cured mice showed T-cell unresponsiveness to InsB9-23 stimulation and increased Treg frequency in islet infiltration and pancreatic lymph nodes. Additional studies revealed a complex mechanism of Ag-specific immune regulation driven by CT25, in which both Tregs and PDL1 costimulation cooperate to control diabetogenic cells, while transplanted islets play a crucial role, although transient, recruiting diabetogenic cells. Therefore, CT25 before alloislet transplantation represents an Ag-specific immunotherapy to resolve autoimmune diabetes in the presence of residual endogenous ß-cell mass.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Hepatocitos/metabolismo , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Péptidos/uso terapéutico , Animales , Autoinmunidad/efectos de los fármacos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Técnicas de Transferencia de Gen , Supervivencia de Injerto/inmunología , Hepatocitos/inmunología , Células Secretoras de Insulina/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Recurrencia , Prevención Secundaria , Linfocitos T Reguladores/inmunología
20.
J Clin Endocrinol Metab ; 106(2): e507-e519, 2021 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-33124663

RESUMEN

AIM: The aim of this study was to investigate whether treatment with rapamycin plus vildagliptin restores ß-cell function in patients with long-standing type 1 diabetes. METHODS: A phase 2, single-center, randomized, double-blind, placebo-controlled study was conducted in long-standing type 1 diabetes patients randomly assigned (1:1:1) to 4 weeks of rapamycin (group 2), 4 weeks of rapamycin plus 12 weeks of vildagliptin (group 3), or double placebo (group 1). The primary outcome was the proportion of participants with a positive response to the Mixed-Meal Tolerance Test (C-peptide at 90 minutes > 0.2 nmol/L) at weeks 4 and 12. Secondary end points included insulin requirement, standard measures of glycemic control, and hormonal and immunological profile. RESULTS: Fifty-five patients were randomly assigned to group 1 (n = 18), group 2 (n = 19), or group 3 (n = 18). No patient in any group showed a positive C-peptide response, and there was no significant difference at 4 and 12 weeks for the primary outcome. At 4 weeks, insulin requirement decreased from 0.54 to 0.48 U/kg/day in group 2 (P = .013), from 0.59 to 0.51 U/kg/day in group 3 (P < .001), whereas it did not change in group 1. At 12 weeks, glycated hemoglobin significantly decreased both in group 2 (from 7.3% [56 mmol/mol] to 7% [53 mmol/mol]; P = .045] and in group 3 (from 7.2% [55.5 mmol/mol] to 6.9% [52 mmol/mol]; P = .001]. Rapamycin treatment was associated with a decrease in insulin antibody titer and changes in hormonal/immunological profile. CONCLUSIONS: Rapamycin reduced insulin requirement, but did not restore ß-cell function in patients with long-standing type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Células Secretoras de Insulina/efectos de los fármacos , Sirolimus/administración & dosificación , Vildagliptina/administración & dosificación , Adulto , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Método Doble Ciego , Quimioterapia Combinada , Femenino , Humanos , Células Secretoras de Insulina/fisiología , Italia , Masculino , Persona de Mediana Edad , Placebos , Recuperación de la Función/efectos de los fármacos , Sirolimus/farmacología , Resultado del Tratamiento , Vildagliptina/farmacología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA