Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cancer Immunol Immunother ; 67(10): 1559-1569, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30056598

RESUMEN

Current treatments for tumors expressing epidermal growth factor receptor (EGFR) include anti-EGFR monoclonal antibodies, often used in conjunction with the standard chemotherapy, radiation therapy, or other EGFR inhibitors. While monoclonal antibody treatment is efficacious in many patients, drawbacks include its high cost of treatment and side effects associated with multiple drug infusions. As an alternative to monoclonal antibody treatments, we have focused on peptide-based vaccination to trigger natural anti-tumor antibodies. Here, we demonstrate that peptides based on a region of the EGFR extracellular domain IV break immune tolerance to EGFR and elicit anti-tumor immunity. Mice immunized with isoforms of EGFR peptide p580-598 generated anti-EGFR antibody and T-cell responses. Iso-aspartyl (iso-Asp)-modified EGFR p580 immune sera inhibit in vitro growth of EGFR overexpressing human A431 tumor cells, as well as promote antibody-dependent cell-mediated cytotoxicity (ADCC). Antibodies induced by Asp and iso-Asp p580 bound homologous regions of the EGFR family members HER2 and HER3. EGFR p580 immune sera also inhibited the growth of the human tumor cell line MDA-MB-453 that expresses HER2 but not EGFR. Asp and iso-Asp EGFR p580 induced antibodies were also able to inhibit the in vivo growth of EGFR-expressing tumors. These data demonstrate that EGFR peptides from a region of the EGFR extracellular domain IV promote anti-tumor immunity, tumor cell killing, and antibodies that are cross reactive with ErbB family members.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias de la Mama/inmunología , Receptores ErbB/inmunología , Fragmentos de Péptidos/inmunología , Receptor ErbB-2/inmunología , Receptor ErbB-3/inmunología , Células A549 , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Receptores ErbB/antagonistas & inhibidores , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fragmentos de Péptidos/administración & dosificación , Fosforilación , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/antagonistas & inhibidores , Transducción de Señal , Células Tumorales Cultivadas , Vacunación
2.
J Immunol ; 193(2): 587-96, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24943216

RESUMEN

Immune-deficient mice, reconstituted with human stem cells, have been used to analyze human immune responses in vivo. Although they have been used to study immune responses to xenografts, allografts, and pathogens, there have not been models of autoimmune disease in which the mechanisms of the pathologic process can be analyzed. We have found that reconstituted "humanized" mice treated with anti-CTLA-4 Ab (ipilimumab) develop autoimmune disease characterized by hepatitis, adrenalitis, sialitis, anti-nuclear Abs, and weight loss. Induction of autoimmunity involved activation of T cells and cytokine production, and increased infiltration of APCs. When anti-CTLA-4 mAb-treated mice were cotreated with anti-CD3 mAb (teplizumab), hepatitis and anti-nuclear Abs were no longer seen and weight loss did not occur. The anti-CD3 blocked proliferation and activation of T cells, release of IFN-γ and TNF, macrophage infiltration, and release of IP-10 that was induced with anti-CTLA-4 mAb. We also found increased levels of T regulatory cells (CD25(+)CD127(-)) in the spleen and mesenteric lymph nodes in the mice treated with both Abs and greater constitutive phosphorylation of STAT5 in T regulatory cells in spleen cells compared with mice treated with anti-CTLA-4 mAb alone. We describe a model of human autoimmune disease in vivo. Humanized mice may be useful for understanding the mechanisms of biologics that are used in patients. Hepatitis, lymphadenopathy, and other inflammatory sequelae are adverse effects of ipilimumab treatment in humans, and this study may provide insights into this pathogenesis and the effects of immunologics on autoimmunity.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Enfermedades Autoinmunes/terapia , Modelos Animales de Enfermedad , Trasplante de Células Madre/métodos , Linfocitos T/inmunología , Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/inmunología , Glándulas Suprarrenales/metabolismo , Animales , Anticuerpos Monoclonales/toxicidad , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Citocinas/sangre , Citocinas/inmunología , Citocinas/metabolismo , Citometría de Flujo , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Ipilimumab , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Factor de Transcripción STAT5/inmunología , Factor de Transcripción STAT5/metabolismo , Análisis de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Heterólogo , Pérdida de Peso/efectos de los fármacos , Pérdida de Peso/inmunología
3.
J Biol Chem ; 287(8): 5310-6, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22215667

RESUMEN

B cell acquisition and presentation of specific autoantigens (auto-Ags) are thought to play an important and complex role in autoimmunity development. We previously identified scavenger receptor A (SR-A) as an early target in altering B cell-mediated autoimmunity. SR-A is highly expressed on professional antigen-presenting cells such as macrophages (MΦs) and dendritic cells (DCs). In this study, we demonstrate that SR-A is responsible for controlling B cell interactions with DCs/MΦs to promote Ag transfer from B cells to DCs/MΦs. We established a high-throughput ELISA-based screen to identify novel SR-A inhibitors, the specificity of which was determined by dose dependence and Biacore surface plasmon resonance testing. We identified small molecule inhibitors (SMIs) able to reduce SR-A-mediated Ag transfer in human cells. In particular, the SMIs prevented SR-A-positive cells from accumulating/loading Ag over time. Furthermore, we determined that one SMI, sennoside B, can reduce SR-A-mediated capture of B cells. Finally, SMI-mediated decreases in Ag transfer or accumulation reduced T cell proliferation in vitro and in vivo. These observations demonstrate that B cell-DC/MΦ interactions are conducive to promoting Ag trafficking between these cell types via SR-A. Inhibitors of SR-A may provide a novel therapeutic strategy in ameliorating autoimmune disease development.


Asunto(s)
Antígenos/metabolismo , Receptores Depuradores de Clase A/antagonistas & inhibidores , Receptores Depuradores de Clase A/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antraquinonas/farmacología , Antígenos/inmunología , Autoantígenos/inmunología , Autoantígenos/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Transporte Biológico/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Extracto de Senna , Senósidos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo
4.
Autoimmunity ; 56(1): 2282945, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37994408

RESUMEN

Protein posttranslational modifications (PTMs) arise in a number of normal cellular biological pathways and in response to pathology caused by inflammation and/or infection. Indeed, a number of PTMs have been identified and linked to specific autoimmune responses and metabolic pathways. One particular PTM, termed isoaspartyl (isoAsp or isoD) modification, is among the most common spontaneous PTM occurring at physiological pH and temperature. Herein, we demonstrate that isoAsp modifications arise within the ZAP70 protein tyrosine kinase upon T-cell antigen receptor (TCR) engagement. The enzyme protein L-isoaspartate O-methyltransferase (PCMT1, or PIMT, EC 2.1.1.77) evolved to repair isoaspartyl modifications in cells. In this regard, we observe that increased levels of isoAsp modification that arise under oxidative stress are correlated with reduced PIMT activity in patients with systemic lupus erythematosus (SLE). PIMT deficiency leads to T cell hyper-proliferation and hyper-phosphorylation through ZAP70 signaling. We demonstrate that inducing the overexpression of PIMT can correct the hyper-responsive phenotype in lupus T cells. Our studies reveal a phenotypic role of isoAsp modification and phosphorylation of ZAP70 in lupus T cell autoimmunity and provide a potential therapeutic target through the repair of isoAsp modification.


Asunto(s)
Proteína D-Aspartato-L-Isoaspartato Metiltransferasa , Linfocitos T , Humanos , Linfocitos T/metabolismo , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/genética , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/metabolismo , Estrés Oxidativo , Autoinmunidad , Procesamiento Proteico-Postraduccional , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/metabolismo
5.
EBioMedicine ; 87: 104379, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36463755

RESUMEN

BACKGROUND: Stress responses within the ß cell have been linked with both increased ß cell death and accelerated immune activation in type 1 diabetes (T1D). At present, information on the timing and scope of these responses as well as disease-related changes in islet ß cell protein expression during T1D development is lacking. METHODS: Data independent acquisition-mass spectrometry was performed on islets collected longitudinally from NOD mice and NOD-SCID mice rendered diabetic through T cell adoptive transfer. FINDINGS: In islets collected from female NOD mice at 10, 12, and 14 weeks of age, we found a time-restricted upregulation of proteins involved in stress mitigation and maintenance of ß cell function, followed by loss of expression of protective proteins that heralded diabetes onset. EIF2 signalling and the unfolded protein response, mTOR signalling, mitochondrial function, and oxidative phosphorylation were commonly modulated pathways in both NOD mice and NOD-SCID mice rendered acutely diabetic by T cell adoptive transfer. Protein disulphide isomerase A1 (PDIA1) was upregulated in NOD islets and pancreatic sections from human organ donors with autoantibody positivity or T1D. Moreover, PDIA1 plasma levels were increased in pre-diabetic NOD mice and in the serum of children with recent-onset T1D compared to non-diabetic controls. INTERPRETATION: We identified a core set of modulated pathways across distinct mouse models of T1D and identified PDIA1 as a potential human biomarker of ß cell stress in T1D. FUNDING: NIH (R01DK093954, DK127308, U01DK127786, UC4DK104166, R01DK060581, R01GM118470, and 5T32DK101001-09). VA Merit Award I01BX001733. JDRF (2-SRA-2019-834-S-B, 2-SRA-2018-493-A-B, 3-PDF-20016-199-A-N, 5-CDA-2022-1176-A-N, and 3-PDF-2017-385-A-N).


Asunto(s)
Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Animales , Niño , Femenino , Humanos , Ratones , Biomarcadores/metabolismo , Islotes Pancreáticos/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Proteína Disulfuro Isomerasas/metabolismo , Proteómica , Células Secretoras de Insulina
6.
Front Immunol ; 13: 1028130, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36389721

RESUMEN

Posttranslational protein modifications (PTMs) are an inherent response to physiological changes causing altered protein structure and potentially modulating important biological functions of the modified protein. Besides cellular metabolic pathways that may be dictated by PTMs, the subtle change of proteins also may provoke immune attack in numerous autoimmune diseases. Type 1 diabetes (T1D) is a chronic autoimmune disease destroying insulin-producing beta cells within the pancreatic islets, a result of tissue inflammation to specific autoantigens. This review summarizes how PTMs arise and the potential pathological consequence of PTMs, with particular focus on specific autoimmunity to pancreatic beta cells and cellular metabolic dysfunction in T1D. Moreover, we review PTM-associated biomarkers in the prediction, diagnosis and in monitoring disease activity in T1D. Finally, we will discuss potential preventive and therapeutic approaches of targeting PTMs in repairing or restoring normal metabolic pathways in pancreatic islets.


Asunto(s)
Enfermedades Autoinmunes , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Humanos , Autoinmunidad , Enfermedades Autoinmunes/metabolismo , Biomarcadores/metabolismo
7.
Diabetes ; 71(9): 1979-1993, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35730902

RESUMEN

Inflammation and oxidative stress in pancreatic islets amplify the appearance of various posttranslational modifications to self-proteins. In this study, we identified a select group of carbonylated islet proteins arising before the onset of hyperglycemia in NOD mice. Of interest, we identified carbonyl modification of the prolyl-4-hydroxylase ß subunit (P4Hb) that is responsible for proinsulin folding and trafficking as an autoantigen in both human and murine type 1 diabetes. We found that carbonylated P4Hb is amplified in stressed islets coincident with decreased glucose-stimulated insulin secretion and altered proinsulin-to-insulin ratios. Autoantibodies against P4Hb were detected in prediabetic NOD mice and in early human type 1 diabetes prior to the onset of anti-insulin autoimmunity. Moreover, we identify autoreactive CD4+ T-cell responses toward carbonyl-P4Hb epitopes in the circulation of patients with type 1 diabetes. Our studies provide mechanistic insight into the pathways of proinsulin metabolism and in creating autoantigenic forms of insulin in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Animales , Autoantígenos , Autoinmunidad , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Proinsulina/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo
8.
Nat Commun ; 13(1): 1870, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35388005

RESUMEN

Inflammation, including reactive oxygen species and inflammatory cytokines in tissues amplify various post-translational modifications of self-proteins. A number of post-translational modifications have been identified as autoimmune biomarkers in the initiation and progression of Type 1 diabetes. Here we show the citrullination of pancreatic glucokinase as a result of inflammation, triggering autoimmunity and affecting glucokinase biological functions. Glucokinase is expressed in hepatocytes to regulate glycogen synthesis, and in pancreatic beta cells as a glucose sensor to initiate glycolysis and insulin signaling. We identify autoantibodies and autoreactive CD4+ T cells to glucokinase epitopes in the circulation of Type 1 diabetes patients and NOD mice. Finally, citrullination alters glucokinase biologic activity and suppresses glucose-stimulated insulin secretion. Our study define glucokinase as a Type 1 diabetes biomarker, providing new insights of how inflammation drives post-translational modifications to create both neoautoantigens and affect beta cell metabolism.


Asunto(s)
Diabetes Mellitus Tipo 1 , Glucoquinasa , Animales , Citrulinación , Diabetes Mellitus Tipo 1/metabolismo , Glucoquinasa/genética , Glucosa/metabolismo , Humanos , Inflamación/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos NOD
9.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35925682

RESUMEN

Checkpoint inhibitors (CPIs) targeting programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have revolutionized cancer treatment but can trigger autoimmune complications, including CPI-induced diabetes mellitus (CPI-DM), which occurs preferentially with PD-1 blockade. We found evidence of pancreatic inflammation in patients with CPI-DM with shrinkage of pancreases, increased pancreatic enzymes, and in a case from a patient who died with CPI-DM, peri-islet lymphocytic infiltration. In the NOD mouse model, anti-PD-L1 but not anti-CTLA-4 induced diabetes rapidly. RNA sequencing revealed that cytolytic IFN-γ+CD8+ T cells infiltrated islets with anti-PD-L1. Changes in ß cells were predominantly driven by IFN-γ and TNF-α and included induction of a potentially novel ß cell population with transcriptional changes suggesting dedifferentiation. IFN-γ increased checkpoint ligand expression and activated apoptosis pathways in human ß cells in vitro. Treatment with anti-IFN-γ and anti-TNF-α prevented CPI-DM in anti-PD-L1-treated NOD mice. CPIs targeting the PD-1/PD-L1 pathway resulted in transcriptional changes in ß cells and immune infiltrates that may lead to the development of diabetes. Inhibition of inflammatory cytokines can prevent CPI-DM, suggesting a strategy for clinical application to prevent this complication.


Asunto(s)
Diabetes Mellitus , Receptor de Muerte Celular Programada 1 , Animales , Humanos , Mediadores de Inflamación , Ratones , Ratones Endogámicos NOD , Inhibidores del Factor de Necrosis Tumoral
10.
J Exp Med ; 202(9): 1171-7, 2005 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-16260486

RESUMEN

Previous studies (Leadbetter, E.A., I.R. Rifkin, A.H. Hohlbaum, B. Beaudette, M.J. Shlomchik, and A. Marshak-Rothstein. 2002. Nature. 416:603-607; Viglianti, G.A., C.M. Lau, T.M. Hanley, B.A. Miko, M.J. Shlomchik, and A. Marshak-Rothstein. 2003. Immunity. 19:837-847) established the unique capacity of DNA and DNA-associated autoantigens to activate autoreactive B cells via sequential engagement of the B cell antigen receptor (BCR) and Toll-like receptor (TLR) 9. We demonstrate that this two-receptor paradigm can be extended to the BCR/TLR7 activation of autoreactive B cells by RNA and RNA-associated autoantigens. These data implicate TLR recognition of endogenous ligands in the response to both DNA- and RNA-associated autoantigens. Importantly, the response to RNA-associated autoantigens was markedly enhanced by IFN-alpha, a cytokine strongly linked to disease progression in patients with systemic lupus erythematosus (SLE). As further evidence that TLRs play a key role in autoantibody responses in SLE, we found that autoimmune-prone mice, lacking the TLR adaptor protein MyD88, had markedly reduced chromatin, Sm, and rheumatoid factor autoantibody titers.


Asunto(s)
Autoantígenos/inmunología , Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/fisiología , ARN/metabolismo , Receptores de Antígenos de Linfocitos B/fisiología , Receptor Toll-Like 7/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos de Diferenciación/genética , Autoanticuerpos/biosíntesis , Autoantígenos/metabolismo , Linfocitos B/metabolismo , Femenino , Hibridomas , Interferón-alfa/fisiología , Activación de Linfocitos/genética , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide , Receptores de Antígenos de Linfocitos B/genética , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Ribonucleoproteínas/inmunología , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/fisiología
11.
Front Immunol ; 12: 678953, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34140951

RESUMEN

The generation of post-translational modifications (PTMs) in human proteins is a physiological process leading to structural and immunologic variety in proteins, with potentially altered biological functions. PTMs often arise through normal responses to cellular stress, including general oxidative changes in the tissue microenvironment and intracellular stress to the endoplasmic reticulum or immune-mediated inflammatory stresses. Many studies have now illustrated the presence of 'neoepitopes' consisting of PTM self-proteins that induce robust autoimmune responses. These pathways of inflammatory neoepitope generation are commonly observed in many autoimmune diseases including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and type 1 diabetes (T1D), among others. This review will focus on one specific PTM to self-proteins known as citrullination. Citrullination is mediated by calcium-dependent peptidylarginine deiminase (PAD) enzymes, which catalyze deimination, the conversion of arginine into the non-classical amino acid citrulline. PADs and citrullinated peptides have been associated with different autoimmune diseases, notably with a prominent role in the diagnosis and pathology of rheumatoid arthritis. More recently, an important role for PADs and citrullinated self-proteins has emerged in T1D. In this review we will provide a comprehensive overview on the pathogenic role for PADs and citrullination in inflammation and autoimmunity, with specific focus on evidence for their role in T1D. The general role of PADs in epigenetic and transcriptional processes, as well as their crucial role in histone citrullination, neutrophil biology and neutrophil extracellular trap (NET) formation will be discussed. The latter is important in view of increasing evidence for a role of neutrophils and NETosis in the pathogenesis of T1D. Further, we will discuss the underlying processes leading to citrullination, the genetic susceptibility factors for increased recognition of citrullinated epitopes by T1D HLA-susceptibility types and provide an overview of reported autoreactive responses against citrullinated epitopes, both of T cells and autoantibodies in T1D patients. Finally, we will discuss recent observations obtained in NOD mice, pointing to prevention of diabetes development through PAD inhibition, and the potential role of PAD inhibitors as novel therapeutic strategy in autoimmunity and in T1D in particular.


Asunto(s)
Autoinmunidad , Citrulinación , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/metabolismo , Susceptibilidad a Enfermedades , Desiminasas de la Arginina Proteica/metabolismo , Animales , Modelos Animales de Enfermedad , Epigenómica , Epítopos/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación/etiología , Inflamación/metabolismo , Isoenzimas/metabolismo , Procesamiento Proteico-Postraduccional , Desiminasas de la Arginina Proteica/genética , Transducción de Señal
12.
Transl Oncol ; 14(11): 101205, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34419682

RESUMEN

Epidermal Growth Factor Receptor (EGFR) is overexpressed on a number of human cancers, and often is indicative of a poor outcome. Treatment of EGFR/HER2 overexpressing cancers includes monoclonal antibody therapy (cetuximab/trastuzumab) either alone or in conjunction with other standard cancer therapies. While monoclonal antibody therapy has been proven to be efficacious in the treatment of EGFR/HER2 overexpressing tumors, drawbacks include the lack of long-lasting immunity and acquired resistance to monoclonal therapy. An alternative approach is to induce a polyclonal anti-EGFR/HER2 tumor antigen response by vaccine therapy. In this phase I/II open-label study, we examined anti-tumor immunity in companion dogs with spontaneous EGFR expressing tumors. Canine cancers represent an outbred population in which the initiation, progression of disease, mutations and growth factors closely resemble that of human cancers. Dogs with EGFR expressing tumors were immunized with a short peptide of the EGFR extracellular domain with sequence homology to HER2. Serial serum analyses demonstrated high titers of EGFR/HER2 binding antibodies with biological activity similar to that of cetuximab and trastuzumab. Canine antibodies bound both canine and human EGFR on tumor cell lines and tumor tissue. CD8 T cells and IgG deposition were evident in tumors from immunized dogs. The antibodies inhibited EGFR intracellular signaling and inhibited tumor growth in vitro. Additionally, we illustrate objective responses in reducing tumors at metastatic sites in host animals. The data support the approach of amplifying anti-tumor immunity that may be relevant in combination with other immune modifying therapies such as checkpoint inhibitors.

13.
Diabetes ; 70(12): 2879-2891, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34561224

RESUMEN

In type 1 diabetes, autoimmune ß-cell destruction may be favored by neoantigens harboring posttranslational modifications (PTMs) such as citrullination. We studied the recognition of native and citrullinated glucose-regulated protein (GRP)78 peptides by CD8+ T cells. Citrullination modulated T-cell recognition and, to a lesser extent, HLA-A2 binding. GRP78-reactive CD8+ T cells circulated at similar frequencies in healthy donors and donors with type 1 diabetes and preferentially recognized either native or citrullinated versions, without cross-reactivity. Rather, the preference for native GRP78 epitopes was associated with CD8+ T cells cross-reactive with bacterial mimotopes. In the pancreas, a dominant GRP78 peptide was instead preferentially recognized when citrullinated. To further clarify these recognition patterns, we considered the possibility of citrullination in the thymus. Citrullinating peptidylarginine deiminase (Padi) enzymes were expressed in murine and human medullary epithelial cells (mTECs), with citrullinated proteins detected in murine mTECs. However, Padi2 and Padi4 expression was diminished in mature mTECs from NOD mice versus C57BL/6 mice. We conclude that, on one hand, the CD8+ T cell preference for native GRP78 peptides may be shaped by cross-reactivity with bacterial mimotopes. On the other hand, PTMs may not invariably favor loss of tolerance because thymic citrullination, although impaired in NOD mice, may drive deletion of citrulline-reactive T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citrulinación/fisiología , Diabetes Mellitus Tipo 1/inmunología , Chaperón BiP del Retículo Endoplásmico/inmunología , Epítopos de Linfocito T/metabolismo , Adolescente , Adulto , Animales , Niño , Citrulinación/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Chaperón BiP del Retículo Endoplásmico/química , Chaperón BiP del Retículo Endoplásmico/metabolismo , Epítopos de Linfocito T/química , Femenino , Humanos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Procesamiento Proteico-Postraduccional/inmunología , Procesamiento Proteico-Postraduccional/fisiología , Adulto Joven
14.
J Immunol ; 181(6): 4043-51, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768860

RESUMEN

B lymphocytes can function independently as efficient APCs. However, our previous studies demonstrate that both dendritic cells and macrophages are necessary to propagate immune responses initiated by B cell APCs. This finding led us to identify a process in mice whereby Ag-specific B cells transfer Ag to other APCs. In this study, we report the ability and mechanism by which human B lymphocytes can transfer BCR-captured Ag to macrophages. The transfer of Ag involves direct contact between the two cells followed by the capture of B cell-derived membrane and/or intracellular components by the macrophage. These events are abrogated by blocking scavenger receptor A, a receptor involved in the exchange of membrane between APCs. Macrophages acquire greater amounts of Ag in the presence of specific B cells than in their absence. This mechanism allows B cells to amplify or edit the immune response to specific Ag by transferring BCR-captured Ag to other professional APCs, thereby increasing the frequency of its presentation. Ag transfer may perpetuate chronic autoimmune responses to specific self-proteins and help explain the efficacy of B cell-directed therapies in human disease.


Asunto(s)
Presentación de Antígeno/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Comunicación Celular/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Receptores Depuradores/fisiología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Membrana Celular/inmunología , Membrana Celular/metabolismo , Técnicas de Cocultivo , Citosol/inmunología , Citosol/metabolismo , Humanos , Transporte de Proteínas/inmunología , Receptores Depuradores/clasificación , Transducción de Señal/inmunología
15.
Front Endocrinol (Lausanne) ; 11: 624590, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33679609

RESUMEN

Due to their secretory function, ß cells are predisposed to higher levels of endoplasmic reticulum (ER) stress and greater sensitivity to inflammation than other cell types. These stresses elicit changes in ß cells that alter their function and immunogenicity, including defective ribosomal initiation, post-translational modifications (PTMs) of endogenous ß cell proteins, and alternative splicing. Multiple published reports confirm the presence of not only CD8+ T cells, but also autoreactive CD4+ T cells within pancreatic islets. Although the specificities of T cells that infiltrate human islets are incompletely characterized, they have been confirmed to include neo-epitopes that are formed through stress-related enzymatic modifications of ß cell proteins. This article summarizes emerging knowledge about stress-induced changes in ß cells and data supporting a role for neo-antigen formation and cross-talk between immune cells and ß cells that provokes autoimmune attack - leading to a breakdown in tissue-specific tolerance in subjects who develop type 1 diabetes.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Epítopos de Linfocito B/inmunología , Estrés Oxidativo/inmunología , Animales , Humanos , Procesamiento Proteico-Postraduccional/inmunología
16.
Diabetes ; 67(6): 1035-1042, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29784651

RESUMEN

Prior to the onset of type 1 diabetes, there is progressive loss of immune self-tolerance, evidenced by the accumulation of islet autoantibodies and emergence of autoreactive T cells. Continued autoimmune activity leads to the destruction of pancreatic ß-cells and loss of insulin secretion. Studies of samples from patients with type 1 diabetes and of murine disease models have generated important insights about genetic and environmental factors that contribute to susceptibility and immune pathways that are important for pathogenesis. However, important unanswered questions remain regarding the events that surround the initial loss of tolerance and subsequent failure of regulatory mechanisms to arrest autoimmunity and preserve functional ß-cells. In this Perspective, we discuss various processes that lead to the generation of neoepitopes in pancreatic ß-cells, their recognition by autoreactive T cells and antibodies, and potential roles for such responses in the pathology of disease. Emerging evidence supports the relevance of neoepitopes generated through processes that are mechanistically linked with ß-cell stress. Together, these observations support a paradigm in which neoepitope generation leads to the activation of pathogenic immune cells that initiate a feed-forward loop that can amplify the antigenic repertoire toward pancreatic ß-cell proteins.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/metabolismo , Células Secretoras de Insulina/inmunología , Modelos Inmunológicos , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/fisiopatología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Progresión de la Enfermedad , Humanos , Tolerancia Inmunológica , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Activación de Linfocitos , Procesamiento Proteico-Postraduccional , Linfocitos T/metabolismo
17.
Antioxid Redox Signal ; 29(14): 1415-1431, 2018 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29088923

RESUMEN

SIGNIFICANCE: Various autoimmune syndromes are characterized by abnormalities found at the level of tissues and cells, as well as by microenvironmental influences, such as reactive oxygen species (ROS), that alter intracellular metabolism and protein expression. Moreover, the convergence of genetic, epigenetic, and even environmental influences can result in B and T lymphocyte autoimmunity and tissue pathology. Recent Advances: This review describes how oxidative stress to cells and tissues may alter post-translational protein modifications, both directly and indirectly, as well as potentially lead to aberrant gene expression. For example, it has been clearly observed in many systems how oxidative stress directly amplifies carbonyl protein modifications. However, ROS also lead to a number of nonenzymatic spontaneous modifications including deamidation and isoaspartate modification as well as to enzyme-mediated citrullination of self-proteins. ROS have direct effects on DNA methylation, leading to influences in gene expression, chromosome inactivation, and the silencing of genetic elements. Finally, ROS can alter many other cellular pathways, including the initiation of apoptosis and NETosis, triggering the release of modified intracellular autoantigens. CRITICAL ISSUES: This review will detail specific post-translational protein modifications, the pathways that control autoimmunity to modified self-proteins, and how products of ROS may be important biomarkers of tissue pathogenesis. FUTURE DIRECTIONS: A clear understanding of the many pathways affected by ROS will lead to potential therapeutic manipulations to alter the onset and/or progression of autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Animales , Metilación de ADN , Humanos , Oxidación-Reducción , Carbonilación Proteica , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
18.
Diabetes ; 67(11): 2337-2348, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30348823

RESUMEN

The ß-cell has become recognized as a central player in the pathogenesis of type 1 diabetes with the generation of neoantigens as potential triggers for breaking immune tolerance. We report that posttranslationally modified glucose-regulated protein 78 (GRP78) is a novel autoantigen in human type 1 diabetes. When human islets were exposed to inflammatory stress induced by interleukin-1ß, tumor necrosis factor-α, and interferon-γ, arginine residue R510 within GRP78 was converted into citrulline, as evidenced by liquid chromatography-tandem mass spectrometry. This conversion, known as citrullination, led to the generation of neoepitopes, which effectively could be presented by HLA-DRB1*04:01 molecules. With the use of HLA-DRB1*04:01 tetramers and ELISA techniques, we demonstrate enhanced antigenicity of citrullinated GRP78 with significantly increased CD4+ T-cell responses and autoantibody titers in patients with type 1 diabetes compared with healthy control subjects. Of note, patients with type 1 diabetes had a predominantly higher percentage of central memory cells and a lower percentage of effector memory cells directed against citrullinated GRP78 compared with the native epitope. These results strongly suggest that citrullination of ß-cell proteins, exemplified here by the citrullination of GRP78, contributes to loss of self-tolerance toward ß-cells in human type 1 diabetes, indicating that ß-cells actively participate in their own demise.


Asunto(s)
Autoantígenos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Proteínas de Choque Térmico/metabolismo , Inflamación/metabolismo , Islotes Pancreáticos/metabolismo , Autoantígenos/inmunología , Citrulinación , Citocinas/farmacología , Diabetes Mellitus Tipo 1/inmunología , Chaperón BiP del Retículo Endoplásmico , Humanos , Inflamación/inmunología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología
19.
Autoimmunity ; 40(2): 131-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17453712

RESUMEN

The development of immune tolerance is dependent on the expression of self-peptides in the thymus and bone marrow during lymphocyte development. However, not all self-antigens are expressed in the thymus, particularly for proteins that become post-translationally modified during other biological processes in a cell. We have found that one such post-translational modification, the spontaneous conversion of an aspartic acid to isoaspartic acid (isoAsp), causes ignored self-antigens to become immunogenic. In order to determine the mechanism for this autoimmune response, pigeon cytochrome c peptide 88-104 (PCC p88-104) was synthesized with and without an isoaspartyl residue. Each form was digested with cathepsin D, an enzyme involved in antigen processing. The products of cathepsin digestion were dramatically different between the two forms of self-protein suggesting that cryptic self-peptides may be revealed to the immune system by natural modifications to self-proteins. This observation also held true if whole PCC protein contained isoaspartyl residues was digested with cathespsin D. Additionally, AND transgenic TCR T cells (recognizing PCC 88-104) proliferated to a greater extent in response to isoaspartyl PCC as compared to the normal form of PCC. These finding demonstrate the importance of post-translational modifications in shaping autoimmune responses in and the development of tolerance to self-proteins.


Asunto(s)
Citocromos c/inmunología , Ácido Isoaspártico/inmunología , Fragmentos de Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Autoinmunidad , Catepsina D/química , Proliferación Celular , Columbidae , Citocromos c/química , Tolerancia Inmunológica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/citología , Linfocitos T/inmunología
20.
Ann N Y Acad Sci ; 1050: 1-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16014515

RESUMEN

Although the immune system has developed mechanisms to distinguish "self" from "non-self," the presence of autoimmune diseases demonstrates that these mechanisms can be bypassed. The posttranslational modification of self-antigens is one way in which "new" antigens are created for which immune tolerance does not exist. We review some of the posttranslationally modified self-antigens associated with autoimmune diseases, how they arise, and how they break immune tolerance.


Asunto(s)
Autoantígenos/genética , Procesamiento Proteico-Postraduccional/inmunología , Animales , Enfermedades Autoinmunes , Autoinmunidad/inmunología , Predicción , Humanos , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA