Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Autoimmun ; 134: 102961, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36470208

RESUMEN

Regulatory T cell (Treg) therapy is a promising strategy to treat inflammatory bowel disease (IBD). Data from animal models has shown that Tregs specific for intestinal antigens are more potent than polyclonal Tregs at inhibiting colitis. Flagellins, the major structural proteins of bacterial flagella, are immunogenic antigens frequently targeted in IBD subjects, leading to the hypothesis that flagellin-specific Tregs could be an effective cell therapy for IBD. We developed a novel chimeric antigen receptor (CAR) specific for flagellin derived from Escherichia coli H18 (FliC). We used this CAR to confer FliC-specificity to human Tregs and investigated their therapeutic potential. FliC-CAR Tregs were activated by recombinant FliC protein but not a control flagellin protein, demonstrating CAR specificity and functionality. In a humanized mouse model, expression of the FliC-CAR drove preferential migration to the colon and expression of the activation marker PD1. In the presence of recombinant FliC protein in vitro, FliC-CAR Tregs were significantly more suppressive than control Tregs and promoted the establishment of colon-derived epithelial cell monolayers. These results demonstrate the potential of FliC-CAR Tregs to treat IBD and more broadly show the therapeutic potential of CARs targeting microbial-derived antigens.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Receptores Quiméricos de Antígenos , Animales , Ratones , Humanos , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Flagelina/metabolismo , Proteínas Recombinantes/metabolismo , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/metabolismo , Linfocitos T Reguladores
2.
Immunology ; 159(2): 231-241, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31713233

RESUMEN

Regulatory T (Treg) cell-specific deletion of a gene of interest is a procedure widely used to study mechanisms controlling Treg development, homeostasis and function. Accordingly, several transgenic mouse lines have been generated that bear the Cre recombinase under control of the Foxp3 promoter either as a random transgene insertion or knocked into the endogenous Foxp3 locus, with the Foxp3YFP-Cre strain of mice being one of the most widely used. In an attempt to generate Treg cells that lacked expression of the insulin receptor (Insr), we crossed Foxp3YFP-Cre mice with Insrfl/fl mice. Using a conventional two-band PCR genotyping method we found that offspring genotypes did not correspond to the expected Mendelian ratios. We therefore developed a quantitative PCR-based genotyping method to investigate possible ectopic recombination outside the Treg lineage. With this method we found that ~50% of the F1 -generation mice showed evidence of ectopic recombination and that ~10% of the F2 -generation mice had germline Cre recombination activity leading to a high frequency of offspring with global Insr deletion. Use of the quantitative PCR genotyping method enabled accurate selection of mice without ectopic recombination and only the desired Treg cell-specific Insr deletion. Our data highlight the need to use genotyping methods that allow for assessment of possible ectopic recombination driven by the Foxp3YFP-Cre allele, particularly when studying genes that are systemically expressed.


Asunto(s)
Proteínas Bacterianas/genética , Factores de Transcripción Forkhead/genética , Integrasas/genética , Proteínas Luminiscentes/genética , Receptor de Insulina/genética , Recombinación Genética , Linfocitos T Reguladores/inmunología , Animales , Proteínas Bacterianas/biosíntesis , Linaje de la Célula , Cruzamientos Genéticos , Genes Reporteros , Genotipo , Integrasas/metabolismo , Proteínas Luminiscentes/biosíntesis , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Reacción en Cadena de la Polimerasa/métodos , Regiones Promotoras Genéticas , Receptor de Insulina/deficiencia , Linfocitos T Reguladores/metabolismo
3.
Diabetologia ; 60(3): 453-463, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27999871

RESUMEN

AIMS/HYPOTHESIS: A contributor to beta cell failure in type 2 diabetes and islet transplants is amyloid formation by aggregation of the beta cell peptide, islet amyloid polypeptide (IAPP). Similar to the proinsulin processing pathway that generates insulin, IAPP is derived from a prohormone precursor, proIAPP, which requires cleavage by prohormone convertase (PC) 1/3 and PC2 in rodent pancreatic beta cells. We hypothesised that loss of PC2 would promote beta cell death and dysfunction in a rodent model of human beta cell proIAPP overexpression. METHODS: We generated an islet transplant model wherein immune-deficient mouse models of diabetes received islets expressing amyloidogenic human proIAPP and lacking PC2, leading to restoration of normoglycaemia accompanied by increased secretion of human proIAPP. Blood glucose levels were analysed for up to 16 weeks in transplant recipients and grafts were assessed for islet amyloid and beta cell number and death. RESULTS: Hyperglycaemia (blood glucose >16.9 mmol/l) returned in 94% of recipients of islets expressing human proIAPP and lacking PC2, whereas recipients of islets that express human proIAPP and normal PC2 levels remained normoglycaemic for at least 16 weeks. Islet graft failure was accompanied by a ∼20% reduction in insulin-positive cells, yet the degree of amyloid deposition and beta cell apoptosis was similar to those of controls expressing human proIAPP with functional PC2 levels. CONCLUSIONS/INTERPRETATION: PC2 deficiency in transplanted mouse islets expressing human proIAPP promotes beta cell loss and graft failure. Our data suggest that impaired NH2-terminal processing and increased secretion of human proIAPP promote beta cell failure.


Asunto(s)
Amiloide/metabolismo , Células Secretoras de Insulina/metabolismo , Proproteína Convertasa 2/metabolismo , Amiloide/genética , Animales , Glucemia/metabolismo , Western Blotting , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Proinsulina/metabolismo , Proproteína Convertasa 1/genética , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 2/genética
4.
J Immunol ; 194(7): 3054-64, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25740943

RESUMEN

Autoimmune destruction of insulin-producing ß cells in type 1 diabetes and islet transplantation involves a variety of immune pathways but is primarily mediated by self-reactive T cells. Chemokines can modulate local immune responses in inflammation and tumors by recruiting immune cells. We have reported that expression of the chemokine CCL22 in pancreatic ß cells in the NOD mouse prevents autoimmune attack by recruiting T regulatory cells (Tregs), protecting mice from diabetes. In this study we show that invariant NKT cells are also recruited to CCL22-expressing islet transplants and are required for CCL22-mediated protection from autoimmunity. Moreover, CCL22 induces an influx of plasmacytoid dendritic cells, which correlates with higher levels of IDO in CCL22-expressing islet grafts. In addition to its chemotactic properties, we found that CCL22 activates Tregs and promotes their ability to induce expression of IDO by dendritic cells. Islet CCL22 expression thus produces a tolerogenic milieu through the interplay of Tregs, invariant NKT cells, and plasmacytoid dendritic cells, which results in suppression of effector T cell responses and protection of ß cells. The immunomodulatory properties of CCL22 could be harnessed for prevention of graft rejection and type 1 diabetes as well as other autoimmune disorders.


Asunto(s)
Quimiocina CCL22/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Animales , Quimiocina CCL22/genética , Quimiotaxis/genética , Quimiotaxis/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Inmunomodulación/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transducción Genética , Trasplantes/inmunología , Trasplantes/metabolismo
5.
Mol Metab ; 89: 102017, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39182839

RESUMEN

OBJECTIVE: Human embryonic stem cell (hESC; SC)-derived pancreatic ß cells can be used to study diabetes pathologies and develop cell replacement therapies. Although current differentiation protocols yield SCß cells with varying degrees of maturation, these cells still differ from deceased donor human ß cells in several respects. We sought to develop a reporter cell line that could be used to dynamically track SCß cell functional maturation. METHODS: To monitor SCß cell maturation in vitro, we created an IAPP-2A-mScar and INSULIN-2A-EGFP dual fluorescent reporter (INS2A-EGFP/+;IAPP2A-mScarlet/+) hESC line using CRISPR/Cas9. Pluripotent SC were then differentiated using a 7-stage protocol to islet-like cells. Immunohistochemistry, flow cytometry, qPCR, GSIS and electrophysiology were used to characterise resulting cell populations. RESULTS: We observed robust expression of EGFP and mScarlet fluorescent proteins in insulin- and IAPP-expressing cells without any compromise to their differentiation. We show that the proportion of insulin-producing cells expressing IAPP increases over a 4-week maturation period, and that a subset of insulin-expressing cells remain IAPP-free. Compared to this IAPP-free population, we show these insulin- and IAPP-expressing cells are less polyhormonal, more glucose-sensitive, and exhibit decreased action potential firing in low (2.8 mM) glucose. CONCLUSIONS: The INS2A-EGFP/+;IAPP2A-mScarlet/+ hESC line provides a useful tool for tracking populations of maturing hESC-derived ß cells in vitro. This tool has already been shared with 3 groups and is freely available to all.

6.
JCI Insight ; 9(5)2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38319716

RESUMEN

Pattern recognition receptor responses are profoundly attenuated before the third trimester of gestation in the relatively low-oxygen human fetal environment. However, the mechanisms regulating these responses are uncharacterized. Herein, genome-wide transcription and functional metabolic experiments in primary neonatal monocytes linked the negative mTOR regulator DDIT4L to metabolic stress, cellular bioenergetics, and innate immune activity. Using genetically engineered monocytic U937 cells, we confirmed that DDIT4L overexpression altered mitochondrial dynamics, suppressing their activity, and blunted LPS-induced cytokine responses. We also showed that monocyte mitochondrial function is more restrictive in earlier gestation, resembling the phenotype of DDIT4L-overexpressing U937 cells. Gene expression analyses in neonatal granulocytes and lung macrophages in preterm infants confirmed upregulation of the DDIT4L gene in the early postnatal period and also suggested a potential protective role against inflammation-associated chronic neonatal lung disease. Taken together, these data show that DDIT4L regulates mitochondrial activity and provide what we believe to be the first direct evidence for its potential role supressing innate immune activity in myeloid cells during development.


Asunto(s)
Citocinas , Recien Nacido Prematuro , Recién Nacido , Humanos , Citocinas/metabolismo , Monocitos/metabolismo , Inmunidad Innata , Mitocondrias/metabolismo
7.
Mol Metab ; 68: 101667, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36621763

RESUMEN

OBJECTIVES: Pancreatic cancer risk is elevated approximately two-fold in type 1 and type 2 diabetes. Islet amyloid polypeptide (IAPP) is an abundant beta-cell peptide hormone that declines with diabetes progression. IAPP has been reported to act as a tumour-suppressor in p53-deficient cancers capable of regressing tumour volumes. Given the decline of IAPP during diabetes development, we investigated the actions of IAPP in pancreatic ductal adenocarcinoma (PDAC; the most common form of pancreatic cancer) to determine if IAPP loss in diabetes may increase the risk of pancreatic cancer. METHODS: PANC-1, MIA PaCa-2, and H1299 cells were treated with rodent IAPP, and the IAPP analogs pramlintide and davalintide, and assayed for changes in proliferation, death, and glycolysis. An IAPP-deficient mouse model of PDAC (Iapp-/-; Kras+/LSL-G12D; Trp53flox/flox; Ptf1a+/CreER) was generated for survival analysis. RESULTS: IAPP did not impact glycolysis in MIA PaCa-2 cells, and did not impact cell death, proliferation, or glycolysis in PANC-1 cells or in H1299 cells, which were previously reported as IAPP-sensitive. Iapp deletion in Kras+/LSL-G12D; Trp53flox/flox; Ptf1a+/CreER mice had no effect on survival time to lethal tumour burden. CONCLUSIONS: In contrast to previous reports, we find that IAPP does not function as a tumour suppressor. This suggests that loss of IAPP signalling likely does not increase the risk of pancreatic cancer in individuals with diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Neoplasias Pancreáticas , Ratones , Animales , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas
8.
J Clin Invest ; 133(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37561596

RESUMEN

Adoptive immunotherapy with Tregs is a promising approach for preventing or treating type 1 diabetes. Islet antigen-specific Tregs have more potent therapeutic effects than polyclonal cells, but their low frequency is a barrier for clinical application. To generate Tregs that recognize islet antigens, we engineered a chimeric antigen receptor (CAR) derived from a monoclonal antibody with specificity for the insulin B chain 10-23 peptide presented in the context of the IAg7 MHC class II allele present in NOD mice. Peptide specificity of the resulting InsB-g7 CAR was confirmed by tetramer staining and T cell proliferation in response to recombinant or islet-derived peptide. The InsB-g7 CAR redirected NOD Treg specificity such that insulin B 10-23-peptide stimulation enhanced suppressive function, measured via reduction of proliferation and IL-2 production by BDC2.5 T cells and CD80 and CD86 expression on dendritic cells. Cotransfer of InsB-g7 CAR Tregs prevented adoptive transfer diabetes by BDC2.5 T cells in immunodeficient NOD mice. In WT NOD mice, InsB-g7 CAR Tregs prevented spontaneous diabetes. These results show that engineering Treg specificity for islet antigens using a T cell receptor-like CAR is a promising therapeutic approach for the prevention of autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Receptores Quiméricos de Antígenos , Ratones , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/prevención & control , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Ratones Endogámicos NOD , Insulina/metabolismo , Linfocitos T Reguladores
9.
bioRxiv ; 2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36865264

RESUMEN

Adoptive immunotherapy with Tregs is a promising approach for prevention or treatment of type 1 diabetes. Islet antigen-specific Tregs have more potent therapeutic effects than polyclonal cells, but their low frequency is a barrier for clinical application. To generate Tregs that recognize islet antigens, we engineered a chimeric antigen receptor (CAR) derived from a monoclonal antibody with specificity for the insulin B-chain 10-23 peptide presented in the context of the IA g7 MHC class II allele present in NOD mice. Peptide specificity of the resulting InsB-g7 CAR was confirmed by tetramer staining and T cell proliferation in response to recombinant or islet-derived peptide. The InsB-g7 CAR re-directed NOD Treg specificity such that insulin B 10-23-peptide stimulation enhanced suppressive function, measured via reduction of proliferation and IL-2 production by BDC2.5 T cells and CD80 and CD86 expression on dendritic cells. Co-transfer of InsB-g7 CAR Tregs prevented adoptive transfer diabetes by BDC2.5 T cells in immunodeficient NOD mice. In wild type NOD mice, InsB-g7 CAR Tregs stably expressed Foxp3 and prevented spontaneous diabetes. These results show that engineering Treg specificity for islet antigens using a T cell receptor-like CAR is a promising new therapeutic approach for the prevention of autoimmune diabetes. Brief Summary: Chimeric antigen receptor Tregs specific for an insulin B-chain peptide presented by MHC class II prevent autoimmune diabetes.

10.
Immunology ; 136(2): 115-22, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22348589

RESUMEN

T regulatory (Treg) cells are critical for maintaining immune homeostasis and establishing tolerance to foreign, non-pathogenic antigens including those found in commensal bacteria and food. Because of their multiple suppressive mechanisms, Tregs represent a promising strategy for engineering tolerance to self and non-self antigens in chronic inflammatory diseases. Already in clinical trials in the transplantation setting, the question remains whether this therapy would be effective for the treatment of mucosal inflammatory diseases that do not pose an immediate threat to life. In this review we will discuss evidence from both animal models and patients suggesting that Treg therapy would be beneficial in the context of inflammatory bowel disease (IBD). We will examine the role of T-cell versus Treg dysfunction in IBD and discuss the putative antigens that could be potential targets of antigen-directed Treg therapy. Finally, the challenges of using Treg therapy in IBD will be discussed, with a specific emphasis on the role that the microbiota may play in the outcome of this treatment. As Treg therapy becomes a bedside reality in the field of transplantation, there is great hope that it will soon also be deployed in the setting of IBD and ultimately prove more effective than the current non-specific immunosuppressive therapies.


Asunto(s)
Enfermedades Inflamatorias del Intestino/terapia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Modelos Animales de Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones
11.
Curr Opin Organ Transplant ; 17(4): 343-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22790068

RESUMEN

PURPOSE OF REVIEW: There is great hope that cellular therapy with regulatory T cells (Tregs) will be an effective way to induce alloantigen specific tolerance, ultimately allowing for reduction or elimination of nonspecific immunosuppression. In the past, considerable effort was focused on defining the optimal ways to isolate and expand Tregs from peripheral or cord blood. Now that expansion of therapeutically relevant numbers of Tregs is feasible, we need to consider what is going to happen to the cells when they are transferred in vivo. RECENT FINDINGS: For optimal function, Tregs must be able to traffic to the correct location(s) and, despite the presence of immunosuppressive therapy, live long enough to transfer their regulatory function to recipient T cells. Within the Treg pool, there are also functionally specialized subsets, identified by chemokine receptor expression and/or cytokine production, which control their trafficking and relative ability to suppress different types of T helper cells, respectively. Recent findings imply that the plasticity of appropriately obtained populations of Tregs may not be of as great concern as previously suggested. Experimental data have also provided evidence as to how one might design adjunctive treatment that best supports the viability and function of Tregs after transfer. SUMMARY: Knowledge of how Tregs work in transplantation comes from studies that do not recapitulate how these cells will be used in humans. There is a need to develop better preclinical models to study how the in-vivo function of human Tregs can be optimized to ensure they can meet the challenge of inducing transplantation tolerance.


Asunto(s)
Trasplante de Órganos , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Inmunidad Celular , Isoantígenos/inmunología , Linfocitos T Reguladores/trasplante
12.
Front Immunol ; 12: 655027, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34012439

RESUMEN

Newborns are frequently affected by mucocutaneous candidiasis. Th17 cells essentially limit mucosal invasion by commensal Candida spp. Here, we sought to understand the molecular basis for the developmental lack of Th17 cell responses in circulating blood neonatal T cells. Naive cord blood CD4 T cells stimulated in Th17-differentiating conditions inherently produced high levels of the interleukin-22 immunoregulatory cytokine, particularly in the presence of neonatal antigen-presenting cells. A genome-wide transcriptome analysis comparing neonatal and adult naïve CD4 T cells ex vivo revealed major developmental differences in gene networks regulating Small Drosophila Mothers Against Decapentaplegic (SMAD) and Signal Transducer and Activator of Transcription 3 (STAT3) signaling. These changes were functionally validated by experiments showing that the requirement for TGF-ß in human Th17 cell differentiation is age-dependent. Moreover, STAT3 activity was profoundly diminished while overexpression of the STAT3 gene restored Th17 cell differentiation capacity in neonatal T cells. These data reveal that Th17 cell responses are developmentally regulated at the gene expression level in human neonates. These developmental changes may protect newborns against pathological Th17 cell responses, at the same time increasing their susceptibility to mucocutaneous candidiasis.


Asunto(s)
Inmunomodulación , Interleucinas/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Factores de Edad , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citocinas/biosíntesis , Humanos , Recién Nacido , Activación de Linfocitos/inmunología , Factor de Transcripción STAT3/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Interleucina-22
13.
J Exp Med ; 196(10): 1335-46, 2002 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-12438424

RESUMEN

T regulatory (Tr) cells are essential for the induction of peripheral tolerance. Several types of Tr cells exist, including CD4(+) T cells which express CD25 constitutively and suppress immune responses via direct cell-to-cell interactions, and type 1 T regulatory (Tr1) cells, which function via secretion of interleukin (IL)-10 and transforming growth factor (TGF)-beta. The relationship between CD25(+)CD4(+) T cells and Tr1 cells remains unclear. Here, we demonstrate at the clonal level that Tr1 and CD25(+)CD4(+) T cells are two distinct subsets of regulatory cells with different cytokine production profiles. Furthermore, CD25(-)CD4(+) T cells can be rendered anergic by IL-10 and differentiated into Tr1 cells in the absence of CD25(+)CD4(+) T cells. Cloned human CD25(+)CD4(+) T cell populations are heterogeneous and only a subset of clones continues to express high levels of CD25 and is suppressive. The intensity of CD25, cytotoxic T lymphocyte antigen (CTLA)-4, and glucocorticoid-induced tumor necrosis factor (TNF) receptor expression correlates with the suppressive capacity of the T cell clones. None of the CD25(+)CD4(+) T cell clones with suppressive function produce IL-10, but all produce TGF-beta. Suppression mediated by CD25(+)CD4(+) T cell clones is partially dependent on TGF-beta, but not on constitutive high expression of CD25. Together these data indicate that naturally occurring human CD25(+)CD4(+) T cells are distinct from IL-10-producing Tr1 cells.


Asunto(s)
Antígenos CD4/inmunología , Inmunoconjugados , Interleucina-10/biosíntesis , Receptores de Interleucina-2/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Abatacept , Antígenos CD , Antígenos de Diferenciación/inmunología , Antígeno CTLA-4 , Anergia Clonal/inmunología , Células Clonales , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-10/inmunología , Linfocitos T Reguladores/inmunología
14.
Nat Neurosci ; 9(6): 824-31, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699508

RESUMEN

Post-translational modification by the lipid palmitate is crucial for the correct targeting and function of many proteins. Here we show that huntingtin (htt) is normally palmitoylated at cysteine 214, which is essential for its trafficking and function. The palmitoylation and distribution of htt are regulated by the palmitoyl transferase huntingtin interacting protein 14 (HIP14). Expansion of the polyglutamine tract of htt, which causes Huntington disease, results in reduced interaction between mutant htt and HIP14 and consequently in a marked reduction in palmitoylation. Mutation of the palmitoylation site of htt, making it palmitoylation resistant, accelerates inclusion formation and increases neuronal toxicity. Downregulation of HIP14 in mouse neurons expressing wild-type and mutant htt increases inclusion formation, whereas overexpression of HIP14 substantially reduces inclusions. These results suggest that the expansion of the polyglutamine tract in htt results in decreased palmitoylation, which contributes to the formation of inclusion bodies and enhanced neuronal toxicity.


Asunto(s)
Proteínas Portadoras/metabolismo , Corteza Cerebral/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Ácido Palmítico/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos/fisiología , Animales , Animales Recién Nacidos , Células COS , Proteínas Portadoras/genética , Células Cultivadas , Corteza Cerebral/citología , Chlorocebus aethiops , Cisteína/metabolismo , Regulación hacia Abajo/genética , Humanos , Proteína Huntingtina , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Transporte de Proteínas/fisiología , Ratas , Expansión de Repetición de Trinucleótido/genética
15.
Sci Transl Med ; 12(557)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32817364

RESUMEN

Antigen-specific regulatory T cells (Tregs) engineered with chimeric antigen receptors (CARs) are a potent immunosuppressive cellular therapy in multiple disease models and could overcome shortcomings of polyclonal Treg therapy. CAR therapy was initially developed with conventional T cells, which have different signaling requirements than do Tregs To date, most of the CAR Treg studies used second-generation CARs, encoding a CD28 or 4-1BB co-receptor signaling domain and CD3ζ, but it was not known if this CAR design was optimal for Tregs Using a human leukocyte antigen-A2-specific CAR platform and human Tregs, we compared 10 CARs with different co-receptor signaling domains and systematically tested their function and CAR-stimulated gene expression profile. Tregs expressing a CAR encoding CD28wt were markedly superior to all other CARs tested in an in vivo model of graft-versus-host disease. In vitro assays revealed stable expression of Helios and an ability to suppress CD80 expression on dendritic cells as key in vitro predictors of in vivo function. This comprehensive study of CAR signaling domain variants in Tregs can be leveraged to optimize CAR design for use in antigen-specific Treg therapy.


Asunto(s)
Receptores Quiméricos de Antígenos , Antígenos CD28 , Humanos , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Linfocitos T Reguladores
16.
J Exp Med ; 217(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32478834

RESUMEN

Adipose tissue (AT) regulatory T cells (T regs) control inflammation and metabolism. Diet-induced obesity causes hyperinsulinemia and diminishes visceral AT (VAT) T reg number and function, but whether these two phenomena were mechanistically linked was unknown. Using a T reg-specific insulin receptor (Insr) deletion model, we found that diet-induced T reg dysfunction is driven by T reg-intrinsic insulin signaling. Compared with Foxp3cre mice, after 13 wk of high-fat diet, Foxp3creInsrfl/fl mice exhibited improved glucose tolerance and insulin sensitivity, effects associated with lower AT inflammation and increased numbers of ST2+ T regs in brown AT, but not VAT. Similarly, Foxp3creInsrfl/fl mice were protected from the metabolic effects of aging, but surprisingly had reduced VAT T regs and increased VAT inflammation compared with Foxp3cre mice. Thus, in both diet- and aging-associated hyperinsulinemia, excessive Insr signaling in T regs leads to undesirable metabolic outcomes. Ablation of Insr signaling in T regs represents a novel approach to mitigate the detrimental effects of hyperinsulinemia on immunoregulation of metabolic syndrome.


Asunto(s)
Envejecimiento/inmunología , Dieta Alta en Grasa/efectos adversos , Grasa Intraabdominal/inmunología , Síndrome Metabólico/inmunología , Receptor de Insulina/deficiencia , Linfocitos T Reguladores/inmunología , Envejecimiento/genética , Envejecimiento/patología , Animales , Eliminación de Gen , Grasa Intraabdominal/patología , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Ratones , Ratones Transgénicos , Receptor de Insulina/inmunología , Linfocitos T Reguladores/patología
17.
JCI Insight ; 4(6)2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30753169

RESUMEN

Chimeric antigen receptor (CAR) technology can be used to engineer the antigen specificity of regulatory T cells (Tregs) and improve their potency as an adoptive cell therapy in multiple disease models. As synthetic receptors, CARs carry the risk of immunogenicity, particularly when derived from nonhuman antibodies. Using an HLA-A*02:01-specific CAR (A2-CAR) encoding a single-chain variable fragment (Fv) derived from a mouse antibody, we developed a panel of 20 humanized A2-CARs (hA2-CARs). Systematic testing demonstrated variations in expression, and ability to bind HLA-A*02:01 and stimulate human Treg suppression in vitro. In addition, we developed a new method to comprehensively map the alloantigen specificity of CARs, revealing that humanization reduced HLA-A cross-reactivity. In vivo bioluminescence imaging showed rapid trafficking and persistence of hA2-CAR Tregs in A2-expressing allografts, with eventual migration to draining lymph nodes. Adoptive transfer of hA2-CAR Tregs suppressed HLA-A2+ cell-mediated xenogeneic graft-versus-host disease and diminished rejection of human HLA-A2+ skin allografts. These data provide a platform for systematic development and specificity testing of humanized alloantigen-specific CARs that can be used to engineer specificity and homing of therapeutic Tregs.


Asunto(s)
Isoantígenos/inmunología , Isoantígenos/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Aloinjertos , Animales , Femenino , Antígenos HLA-A , Antígeno HLA-A2/inmunología , Humanos , Tolerancia Inmunológica , Inmunoterapia , Inmunoterapia Adoptiva , Células K562 , Ratones , Ratones Transgénicos , Anticuerpos de Cadena Única , Piel/patología , Trasplante de Piel , Inmunología del Trasplante , Trasplante Homólogo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Neuron ; 34(5): 807-20, 2002 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12062026

RESUMEN

Extracellular signal-regulated kinases (ERK1 and 2) are synaptic signaling components necessary for several forms of learning. In mice lacking ERK1, we observe a dramatic enhancement of striatum-dependent long-term memory, which correlates with a facilitation of long-term potentiation in the nucleus accumbens. At the cellular level, we find that ablation of ERK1 results in a stimulus-dependent increase of ERK2 signaling, likely due to its enhanced interaction with the upstream kinase MEK. Consistently, such activity change is responsible for the hypersensitivity of ERK1 mutant mice to the rewarding properties of morphine. Our results reveal an unexpected complexity of ERK-dependent signaling in the brain and a critical regulatory role for ERK1 in the long-term adaptive changes underlying striatum-dependent behavioral plasticity and drug addiction.


Asunto(s)
Cuerpo Estriado/enzimología , Potenciación a Largo Plazo/genética , Memoria/fisiología , Proteínas Quinasas Activadas por Mitógenos/deficiencia , Núcleo Accumbens/enzimología , Terminales Presinápticos/enzimología , Transmisión Sináptica/genética , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/enzimología , Animales , Reacción de Prevención/fisiología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Cuerpo Estriado/anomalías , Cuerpo Estriado/citología , Femenino , Hipocampo/citología , Hipocampo/enzimología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Morfina/farmacología , Actividad Motora/genética , Mutación/genética , Red Nerviosa/anomalías , Red Nerviosa/citología , Red Nerviosa/enzimología , Malformaciones del Sistema Nervioso/enzimología , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Núcleo Accumbens/anomalías , Núcleo Accumbens/citología , Terminales Presinápticos/ultraestructura , Regulación hacia Arriba/genética
19.
J Clin Invest ; 115(11): 3276-84, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16211090

RESUMEN

Little is known about the molecules that control the development and function of CD4+ CD25+ Tregs. Recently, it was shown that the transcription factor FOXP3 is necessary and sufficient for the generation of CD4+ CD25+ Tregs in mice. We investigated the capacity of FOXP3 to drive the generation of suppressive CD4+ CD25+ Tregs in humans. Surprisingly, although ectopic expression of FOXP3 in human CD4+ T cells resulted in induction of hyporesponsiveness and suppression of IL-2 production, it did not lead to acquisition of significant suppressor activity in vitro. Similarly, ectopic expression of FOXP3delta2, an isoform found in human CD4+ CD25+ Tregs that lacks exon 2, also failed to induce the development of suppressor T cells. Moreover, when FOXP3 and FOXP3delta2 were simultaneously overexpressed, although the expression of several Treg-associated cell surface markers was significantly increased, only a modest suppressive activity was induced. These data indicate that in humans, overexpression of FOXP3 alone or together with FOXP3delta2 is not an effective method to generate potent suppressor T cells in vitro and suggest that factors in addition to FOXP3 are required during the process of activation and/or differentiation for the development of bona fide Tregs.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/fisiología , Activación de Linfocitos , Linfocitos T Reguladores/citología , Diferenciación Celular/genética , Células Cultivadas , Citocinas/metabolismo , Regulación hacia Abajo/genética , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Humanos , Células Jurkat , Activación de Linfocitos/genética , Mutación , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
20.
Mol Cell Endocrinol ; 477: 48-56, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29883690

RESUMEN

Overexpression of the X-linked inhibitor of apoptosis (XIAP) prevents islet allograft rejection. We constructed an adeno-associated virus expressing XIAP driven by the rat insulin promoter (dsAAV8-RIP-XIAP) for long-term beta-cell gene expression in vivo. Pancreatic delivery of dsAAV8-RIP-XIAP prevented autoimmune diabetes in 70% of non-obese diabetic (NOD) mice, associated with decreased insulitis. Islets from Balb/c mice transduced with dsAAV8-RIP-XIAP were protected following transplantation into streptozotocin (STZ)-diabetic Bl/6 recipients, associated with decreased graft infiltration. Interestingly, dsAAV8-RIP-XIAP transduction induced expression of lactate dehydrogenase (LDHA) and monocarboxylate transporter 1 (MCT1), two genes normally suppressed in beta cells and involved in production and release of lactate, a metabolite known to suppress local immune responses. Transduction of Balb/c islets with AAV8-RIP-LDHA-MCT1 tended to prolong allograft survival following transplant into STZ-diabetic Bl/6 recipients. These findings suggest that XIAP has therapeutic potential in autoimmune diabetes and raise the possibility that local lactate production may play a role in XIAP-mediated immunomodulation.


Asunto(s)
Aloinjertos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Rechazo de Injerto/prevención & control , Inmunomodulación , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Aloinjertos/efectos de los fármacos , Aloinjertos/metabolismo , Animales , Diabetes Mellitus Tipo 1/patología , Glucosa/farmacología , Rechazo de Injerto/inmunología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunomodulación/efectos de los fármacos , Inyecciones , Insulina/genética , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/biosíntesis , Ratones , Ratones Endogámicos NOD , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ratas , Simportadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA