Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 43(8): 1445-1483, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38499786

RESUMEN

Regulatory T (TREG) cells develop via a program orchestrated by the transcription factor forkhead box protein P3 (FOXP3). Maintenance of the TREG cell lineage relies on sustained FOXP3 transcription via a mechanism involving demethylation of cytosine-phosphate-guanine (CpG)-rich elements at conserved non-coding sequences (CNS) in the FOXP3 locus. This cytosine demethylation is catalyzed by the ten-eleven translocation (TET) family of dioxygenases, and it involves a redox reaction that uses iron (Fe) as an essential cofactor. Here, we establish that human and mouse TREG cells express Fe-regulatory genes, including that encoding ferritin heavy chain (FTH), at relatively high levels compared to conventional T helper cells. We show that FTH expression in TREG cells is essential for immune homeostasis. Mechanistically, FTH supports TET-catalyzed demethylation of CpG-rich sequences CNS1 and 2 in the FOXP3 locus, thereby promoting FOXP3 transcription and TREG cell stability. This process, which is essential for TREG lineage stability and function, limits the severity of autoimmune neuroinflammation and infectious diseases, and favors tumor progression. These findings suggest that the regulation of intracellular iron by FTH is a stable property of TREG cells that supports immune homeostasis and limits the pathological outcomes of immune-mediated inflammation.


Asunto(s)
Apoferritinas , Linfocitos T Reguladores , Animales , Humanos , Ratones , Apoferritinas/genética , Apoferritinas/metabolismo , Linaje de la Célula/genética , Citosina/metabolismo , Factores de Transcripción Forkhead , Hierro/metabolismo
2.
Nat Immunol ; 14(4): 372-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23475182

RESUMEN

Intestinal Peyer's patches are essential lymphoid organs for the generation of T cell-dependent immunoglobulin A (IgA) for gut homeostasis. Through the use of interleukin 17 (IL-17) fate-reporter mice, we found here that endogenous cells of the TH17 subset of helper T cells in lymphoid organs of naive mice 'preferentially' homed to the intestines and were maintained independently of IL-23. In Peyer's patches, such TH17 cells acquired a follicular helper T cell (TFH cell) phenotype and induced the development of IgA-producing germinal center B cells. Mice deficient in TH17 cells failed to generate antigen-specific IgA responses, which provides evidence that TH17 cells are the crucial subset required for the production of high-affinity T cell-dependent IgA.


Asunto(s)
Inmunoglobulina A/inmunología , Ganglios Linfáticos Agregados/inmunología , Células Th17/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Femenino , Centro Germinal/citología , Centro Germinal/inmunología , Inmunoglobulina A/biosíntesis , Inmunoglobulina A Secretora/inmunología , Cambio de Clase de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Ganglios Linfáticos Agregados/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
3.
PLoS Pathog ; 17(8): e1009772, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34352039

RESUMEN

Understanding SARS-CoV-2 evolution and host immunity is critical to control COVID-19 pandemics. At the core is an arms-race between SARS-CoV-2 antibody and angiotensin-converting enzyme 2 (ACE2) recognition, a function of the viral protein spike. Mutations in spike impacting antibody and/or ACE2 binding are appearing worldwide, imposing the need to monitor SARS-CoV2 evolution and dynamics in the population. Determining signatures in SARS-CoV-2 that render the virus resistant to neutralizing antibodies is critical. We engineered 25 spike-pseudotyped lentiviruses containing individual and combined mutations in the spike protein, including all defining mutations in the variants of concern, to identify the effect of single and synergic amino acid substitutions in promoting immune escape. We confirmed that E484K evades antibody neutralization elicited by infection or vaccination, a capacity augmented when complemented by K417N and N501Y mutations. In silico analysis provided an explanation for E484K immune evasion. E484 frequently engages in interactions with antibodies but not with ACE2. Importantly, we identified a novel amino acid of concern, S494, which shares a similar pattern. Using the already circulating mutation S494P, we found that it reduces antibody neutralization of convalescent and post-immunization sera, particularly when combined with E484K and with mutations able to increase binding to ACE2, such as N501Y. Our analysis of synergic mutations provides a signature for hotspots for immune evasion and for targets of therapies, vaccines and diagnostics.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , COVID-19/virología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Sustitución de Aminoácidos/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Línea Celular , Humanos , Evasión Inmune , Mutación/genética , Unión Proteica , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
4.
J Immunol ; 206(5): 978-986, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33472908

RESUMEN

The thymus produces precursors of both conventional T cells (Tconv; also known as effector T cells) and regulatory T cells (Treg) whose interactions prevent autoimmunity while allowing efficient protective immune responses. Tumors express a composite of self-antigens and tumor-specific Ags and engage both Tconv and Treg. Along the aging process, the thymus involutes, and tumor prevalence increases, a correlation proposed previously to result from effector cell decline. In this work, we directly tested whether interruption of thymic activity in adult mice affects Foxp3-expressing Treg composition and function and alters tumor immune surveillance. Young adult mice, on two different genetic backgrounds, were surgically thymectomized (TxT) and analyzed or challenged 2 mo later. Cellular analysis revealed a 10-fold decrease in both Tconv and Treg numbers and a bias for activated cells. The persisting Treg displayed reduced stability of Foxp3 expression and, as a population, showed a compromised return to homeostasis upon induced perturbations. We next tested the growth of three tumor models from different tissue origins and/or presenting distinct degrees of spontaneous immunogenicity. In none of these conditions, adult TxT facilitated tumor growth. Rather, TxT enhanced the efficacy of antitumor immunotherapies targeting Treg and/or the immune checkpoint CTLA4, as evidenced by the increased frequency of responder mice and decreased intratumoral Treg to CD8+IFN-γ+ cell ratio. Together, our findings point to a scenario in which abrogation of thymic activities affects preferentially the regulatory over the ridding arm of the immune activities elicited by tumors and argues that higher prevalence of tumors with age cannot be solely attributed to thymic output decline.


Asunto(s)
Neoplasias/inmunología , Neoplasias/terapia , Timo/inmunología , Animales , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Homeostasis/inmunología , Inmunoterapia/métodos , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología , Timectomía/métodos
5.
Eur J Immunol ; 50(3): 439-444, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31729760

RESUMEN

It is well established that therapeutic impairment of Foxp3+ Treg in mice and humans favors immune rejection of solid tumors. Less explored is the impact Foxp3 allelic variants may have on tumor incidence, progression and therapy. In this work, we tested and demonstrate that the Foxp3fgfp reporter allele, found previously to either enhance or reduce Treg function in specific autoimmunity settings, confers increased anti-tumor immunity. Our conclusions stem out of the analysis of three tumor models of different tissue origin, in two murine genetic backgrounds. When compared to wild type animals, mice carrying the Foxp3fgfp allele spontaneously delay, reduce or prevent primary tumor growth, decrease metastasis growth, and potentiate the response to anti-CTLA4 monotherapy. These findings suggest allelic variances at the Foxp3 locus may serve as predictive indicators for personalized therapy and prognostics, and point at possible new therapeutic targets.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Vigilancia Inmunológica/genética , Neoplasias Experimentales/inmunología , Alelos , Animales , Factores de Transcripción Forkhead/genética , Vigilancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología
6.
Biotechnol Bioeng ; 118(6): 2202-2219, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33624859

RESUMEN

Serological assays are valuable tools to study SARS-CoV-2 spread and, importantly, to identify individuals that were already infected and would be potentially immune to a virus reinfection. SARS-CoV-2 Spike protein and its receptor binding domain (RBD) are the antigens with higher potential to develop SARS-CoV-2 serological assays. Moreover, structural studies of these antigens are key to understand the molecular basis for Spike interaction with angiotensin converting enzyme 2 receptor, hopefully enabling the development of COVID-19 therapeutics. Thus, it is urgent that significant amounts of this protein became available at the highest quality. In this study, we produced Spike and RBD in two human derived cell hosts: HEK293-E6 and Expi293F™. We evaluated the impact of different and scalable bioprocessing approaches on Spike and RBD production yields and, more importantly, on these antigens' quality attributes. Using negative and positive sera collected from human donors, we show an excellent performance of the produced antigens, assessed in serologic enzyme-linked immunosorbent assay (ELISA) tests, as denoted by the high specificity and sensitivity of the test. We show robust Spike productions with final yields of approx. 2 mg/L of culture that were maintained independently of the production scale or cell culture strategy. To the best of our knowledge, the final yield of 90 mg/L of culture obtained for RBD production, was the highest reported to date. An in-depth characterization of SARS-CoV-2 Spike and RBD proteins was performed, namely the antigen's oligomeric state, glycosylation profiles, and thermal stability during storage. The correlation of these quality attributes with ELISA performance show equivalent reactivity to SARS-CoV-2 positive serum, for all Spike and RBD produced, and for all storage conditions tested. Overall, we provide straightforward protocols to produce high-quality SARS-CoV-2 Spike and RBD antigens, that can be easily adapted to both academic and industrial settings; and integrate, for the first time, studies on the impact of bioprocess with an in-depth characterization of these proteins, correlating antigen's glycosylation and biophysical attributes to performance of COVID-19 serologic tests.


Asunto(s)
Antígenos Virales/biosíntesis , Glicosilación , Glicoproteína de la Espiga del Coronavirus/biosíntesis , Frío , Ensayo de Inmunoadsorción Enzimática/normas , Congelación , Células HEK293 , Humanos , Conformación Proteica , Estabilidad Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/normas , SARS-CoV-2 , Pruebas Serológicas/normas , Glicoproteína de la Espiga del Coronavirus/normas
7.
J Immunol ; 200(1): 101-109, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29167234

RESUMEN

It has been shown that dominant tolerance, namely in transplantation, requires Foxp3+ regulatory T cells. Although most tolerance-inducing regimens rely on regulatory T cells, we found that induction of tolerance to proteins in aluminum hydroxide can be achieved in Foxp3-deficient mice using nondepleting anti-CD4 Abs. This type of tolerance is Ag specific, and tolerant mice retain immune competence to respond to unrelated Ags. We demonstrated with chicken OVA-specific TCR-transgenic mice that the same tolerizing protocol (CD4 blockade) and the same target Ag (OVA) achieves Foxp3-dependent transplantation tolerance to OVA-expressing skin grafts, but Foxp3-independent tolerance when the Ag is provided as OVA-aluminum hydroxide. In the latter case, we found that tolerance induction triggered recessive mechanisms leading to elimination of effector cells and, simultaneously, a dominant mechanism associated with the emergence of an anergic and regulatory CTLA-4+IL-2lowFoxp3- T cell population, where the tolerance state is IL-10 dependent. Such Foxp3-independent mechanisms can improve the efficacy of tolerance-inducing protocols.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Tolerancia Inmunológica , Trasplante de Piel , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Hidróxido de Aluminio/inmunología , Animales , Anticuerpos/metabolismo , Presentación de Antígeno , Antígenos CD4/inmunología , Diferenciación Celular , Células Cultivadas , Selección Clonal Mediada por Antígenos , Factores de Transcripción Forkhead/genética , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Activación de Linfocitos , Ratones , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/genética , Ovalbúmina/inmunología
8.
Proc Natl Acad Sci U S A ; 114(12): E2420-E2429, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28255084

RESUMEN

In recent years, the myocardium has been rediscovered under the lenses of immunology, and lymphocytes have been implicated in the pathogenesis of cardiomyopathies with different etiologies. Aging is an important risk factor for heart diseases, and it also has impact on the immune system. Thus, we sought to determine whether immunological activity would influence myocardial structure and function in elderly mice. Morphological, functional, and molecular analyses revealed that the age-related myocardial impairment occurs in parallel with shifts in the composition of tissue-resident leukocytes and with an accumulation of activated CD4+ Foxp3- (forkhead box P3) IFN-γ+ T cells in the heart-draining lymph nodes. A comprehensive characterization of different aged immune-deficient mouse strains revealed that T cells significantly contribute to age-related myocardial inflammation and functional decline. Upon adoptive cell transfer, the T cells isolated from the mediastinal lymph node (med-LN) of aged animals exhibited increased cardiotropism, compared with cells purified from young donors or from other irrelevant sites. Nevertheless, these cells caused rather mild effects on cardiac functionality, indicating that myocardial aging might stem from a combination of intrinsic and extrinsic (immunological) factors. Taken together, the data herein presented indicate that heart-directed immune responses may spontaneously arise in the elderly, even in the absence of a clear tissue damage or concomitant infection. These observations might shed new light on the emerging role of T cells in myocardial diseases, which primarily affect the elderly population.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/inmunología , Miocardio/inmunología , Traslado Adoptivo , Animales , Corazón/crecimiento & desarrollo , Humanos , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Allergy ; 74(10): 1920-1933, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30937919

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is one of the most common skin diseases with a multifactorial etiology. Mutations leading to loss of skin barrier function are associated with the development of AD with group 2 innate lymphoid cells (ILC2) promoting acute skin inflammation. Filaggrin-mutant (Flgft/ft ) mice develop spontaneous skin inflammation accompanied by an increase in skin ILC2 numbers, IL-1ß production, and other cytokines recapitulating human AD. Here, we investigated the role of ILC2, effector cytokines, inflammasome activation, and mast cell function on the development of chronic AD-like inflammation in mice. METHODS: Mice with a frameshift mutation in the filaggrin gene develop spontaneous dermatitis. Flgft/ft mice were crossed to cell- or cytokine-deficient mouse strains, or bred under germ-free conditions. Skin inflammation was scored, and microbiome composition was analyzed. Skin protein expression was measured by multiplex immunoassay. Infiltrating cells were analyzed by flow cytometry. RESULTS: Wild-type and Flgft/ft mice significantly differ in their microbiome composition. Furthermore, mutant mice do not develop skin inflammation under germ-free conditions. ILC2 deficiency did not ameliorate chronic dermatitis in Flgft/ft mice, which was also independent of IL-4, IL-5, IL-9, IL-13, IL-17A, and IL-22. Inflammation was independent of NLRP3 inflammasome activation but required IL-1ß and IL-1R1-signaling. Mechanistically, IL-1ß promoted hyperactivation of IL-1R1-expressing mast cells. Treatment with anti-IL-1ß-antibody alleviated dermatitis exacerbation, while antibiotic intervention ameliorated dermatitis in neonatal mice but not in adults with established inflammation. CONCLUSIONS: In summary, we identified a critical role for the microbiome and IL-1ß mediating chronic inflammation in mice with an impaired skin barrier.


Asunto(s)
Dermatitis Atópica/inmunología , Dermatitis Atópica/metabolismo , Inmunidad Innata , Interleucina-1beta/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Animales , Biopsia , Citocinas/metabolismo , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Proteínas Filagrina , Inflamasomas/metabolismo , Linfocitos/patología , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones , Ratones Transgénicos , Microbiota , Fenotipo , Transducción de Señal , Piel/inmunología , Piel/metabolismo , Piel/patología
10.
Am J Physiol Heart Circ Physiol ; 315(5): H1358-H1367, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30095974

RESUMEN

Heart-specific antibodies have been widely associated with myocardial infarction (MI). However, it remains unclear whether autoantibodies mediate disease progression or are a byproduct of cardiac injury. To disambiguate the role of immunoglobulins in MI, we characterized the development of ischemic heart failure in agammaglobulinemic mice (AID-/-µS-/-). Although these animals can produce functional B cells, they cannot synthesize secretory IgM (µS-/-) or perform Ig class switching (AID-/-), leading to complete antibody deficiency. Agammaglobulinemia did not affect overall post-MI survival but resulted in a significant reduction in infarct size. Echocardiographic analyses showed that, compared with wild-type infarcted control mice, AID-/-µS-/- mice exhibited improved cardiac function and reduced remodeling on day 56 post-MI. These differences remained significant even after animals with matched infarct sizes were compared. Infarcted AID-/-µS-/- mice also showed reduced myocardial expression levels of transcripts known to promote adverse remodeling, such as matrix metalloproteinase-9, collagen type I a1, collagen type III a1, and IL-6. An unbiased screening of the heart reactivity potential in the plasma of wild-type MI animals revealed the presence of antibodies that target the myocardial scar and collagenase-sensitive epitopes. Moreover, we found that IgG accumulated within the scar tissues of infarcted mice and remained in close proximity with cells expressing Fcγ receptors (CD16/32), suggesting the existence of an in situ IgG-Fcγ receptor axis. Collectively, our study results confirm that antibodies contribute to ischemic heart failure progression and provide novel insights into the mechanisms underlying this phenomenon. NEW & NOTEWORTHY Our study sheds some light on the long-standing debate over the relevance of autoantibodies in heart failure and might stimulate future research in the field. The observation of extracellular matrix-specific antibodies and the detection of Fcγ receptor-expressing cells within the scar provide novel insights into the mechanisms by which antibodies may contribute to adverse remodeling.


Asunto(s)
Agammaglobulinemia/inmunología , Autoanticuerpos/inmunología , Insuficiencia Cardíaca/prevención & control , Cambio de Clase de Inmunoglobulina , Inmunoglobulina M/inmunología , Infarto del Miocardio/inmunología , Miocarditis/prevención & control , Miocardio/inmunología , Agammaglobulinemia/complicaciones , Agammaglobulinemia/genética , Agammaglobulinemia/metabolismo , Animales , Autoanticuerpos/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Cambio de Clase de Inmunoglobulina/genética , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocarditis/inmunología , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/metabolismo , Miocardio/patología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular
11.
PLoS Genet ; 10(3): e1004182, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24603313

RESUMEN

The accumulation of adaptive mutations is essential for survival in novel environments. However, in clonal populations with a high mutational supply, the power of natural selection is expected to be limited. This is due to clonal interference--the competition of clones carrying different beneficial mutations--which leads to the loss of many small effect mutations and fixation of large effect ones. If interference is abundant, then mechanisms for horizontal transfer of genes, which allow the immediate combination of beneficial alleles in a single background, are expected to evolve. However, the relevance of interference in natural complex environments, such as the gut, is poorly known. To address this issue, we have developed an experimental system which allows to uncover the nature of the adaptive process as Escherichia coli adapts to the mouse gut. This system shows the invasion of beneficial mutations in the bacterial populations and demonstrates the pervasiveness of clonal interference. The observed dynamics of change in frequency of beneficial mutations are consistent with soft sweeps, where different adaptive mutations with similar phenotypes, arise repeatedly on different haplotypes without reaching fixation. Despite the complexity of this ecosystem, the genetic basis of the adaptive mutations revealed a striking parallelism in independently evolving populations. This was mainly characterized by the insertion of transposable elements in both coding and regulatory regions of a few genes. Interestingly, in most populations we observed a complete phenotypic sweep without loss of genetic variation. The intense clonal interference during adaptation to the gut environment, here demonstrated, may be important for our understanding of the levels of strain diversity of E. coli inhabiting the human gut microbiota and of its recombination rate.


Asunto(s)
Adaptación Fisiológica/genética , Escherichia coli/crecimiento & desarrollo , Selección Genética/genética , Estómago/microbiología , Alelos , Animales , Escherichia coli/genética , Escherichia coli/patogenicidad , Variación Genética , Humanos , Ratones , Modelos Genéticos , Mutación
12.
J Am Soc Nephrol ; 27(9): 2748-61, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26825533

RESUMEN

IgA1 mesangial deposition is the hallmark of IgA nephropathy and Henoch-Schönlein purpura, the onset of which often follows infections. Deposited IgA has been reported as polymeric, J chain associated, and often, hypogalactosylated but with no information concerning the influence of the IgA repertoire or the link between immune stimuli and IgA structure. We explored these issues in the α1KI mouse model, which produces polyclonal human IgA1 prone to mesangial deposition. Compared with mice challenged by a conventional environment, mice in a specific pathogen-free environment had less IgA deposition. However, serum IgA of specific pathogen-free mice showed more galactosylation and much lower polymerization. Notably, wild-type, α1KI, and even J chain-deficient mice showed increased polymeric serum IgA on exposure to pathogens. Strict germfree conditions delayed but did not completely prevent deposition; mice housed in these conditions had very low serum IgA levels and produced essentially monomeric IgA. Finally, comparing monoclonal IgA1 that had different variable regions and mesangial deposition patterns indicated that, independently of glycosylation and polymerization, deposition might also depend on IgA carrying specific variable domains. Together with IgA quantities and constant region post-translational modifications, repertoire changes during immune responses might, thus, modulate IgA propensity to deposition. These IgA features are not associated with circulating immune complexes and C3 deposition and are more pertinent to an initial IgA deposition step preceding overt clinical symptoms in patients.


Asunto(s)
Mesangio Glomerular/metabolismo , Inmunoglobulina A/metabolismo , Animales , Formación de Anticuerpos , Femenino , Inmunoglobulina A/inmunología , Masculino , Ratones , Conformación Proteica
13.
J Immunol ; 192(9): 4210-20, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24663679

RESUMEN

The biologically active form of vitamin D, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], is able to promote the generation of tolerogenic mature dendritic cells (mDCs) with an impaired ability to activate autoreactive T cells. These cells could represent a reliable tool for the promotion or restoration of Ag-specific tolerance through vaccination strategies, for example in type 1 diabetes patients. However, successful transfer of 1,25(OH)2D3-treated mDCs (1,25D3-mDCs) depends on the capacity of 1,25(OH)2D3 to imprint a similar tolerogenic profile in cells derived from diabetes-prone donors as from diabetes-resistant donors. In this study, we examined the impact of 1,25(OH)2D3 on the function and phenotype of mDCs originating from healthy (C57BL/6) and diabetes-prone (NOD) mice. We show that 1,25(OH)2D3 is able to imprint a phenotypic tolerogenic profile on DCs derived from both mouse strains. Both NOD- and C57BL/6-derived 1,25D3-mDCs decreased the proliferation and activation of autoreactive T cells in vitro, despite strain differences in the regulation of cytokine/chemokine expression. In addition, 1,25D3-mDCs from diabetes-prone mice expanded CD25(+)Foxp3(+) regulatory T cells and induced intracellular IL-10 production by T cells in vitro. Furthermore, 1,25D3-mDCs exhibited an intact functional migratory capacity in vivo that favors homing to the liver and pancreas of adult NOD mice. More importantly, when cotransferred with activated CD4(+) T cells into NOD.SCID recipients, 1,25D3-mDCs potently dampened the proliferation of autoreactive donor T cells in the pancreatic draining lymph nodes. Altogether, these results argue for the potential of 1,25D3-mDCs to restore Ag-specific immune tolerance and arrest autoimmune disease progression in vivo.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Vitamina D/análogos & derivados , Animales , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Vitamina D/farmacología
14.
Proc Natl Acad Sci U S A ; 110(16): 6494-9, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23576744

RESUMEN

Most Forkhead box P3(+) (Foxp3(+)) CD4 regulatory T cell (Treg) precursors are newly formed thymocytes that acquire Foxp3 expression on antigen encounter in the thymus. Differentiation of Treg, however, can also occur in the periphery. What limits this second layer of self- and nonself-reactive Treg production in physiological conditions remains to be understood. In this work, we tested the hypothesis that, similarly to thymic Treg, the precursors of peripheral Treg are immature T cells. We show that CD4(+)CD8(-)Foxp3(-) thymocytes and recent thymic emigrants (RTEs), contrarily to peripheral naïve mature cells, efficiently differentiate into Treg on transfer into lymphopenic mice. By varying donor and recipient mice and conducting ex vivo assays, we document that the preferential conversion of newly formed T cells does not require intrathymic preactivation, is cell-intrinsic, and correlates with low and high sensitivity to natural inhibitors and inducers of Foxp3 expression, such as IL-6, T-cell receptor triggering, and TGF-ß. Finally, ex vivo analysis of human thymocytes and peripheral blood T cells revealed that human RTE and newly developed T cells share an increased potential to acquire a FOXP3(bright)CD25(high) Treg phenotype. Our findings indicating that RTEs are the precursors of Tregs differentiated in the periphery should guide the design of Treg-based therapies.


Asunto(s)
Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Células Precursoras de Linfocitos T/citología , Linfocitos T Reguladores/citología , Timocitos/citología , Animales , Células Cultivadas , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología , Estadísticas no Paramétricas , Timocitos/inmunología , Timocitos/trasplante , Factor de Crecimiento Transformador beta/inmunología
16.
Blood ; 122(2): 209-18, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23723451

RESUMEN

B-cell numbers and immunoglobulin (Ig) titers can increase several logs during immune responses. In contrast to this plasticity and despite constant renewal, B-cell numbers are stable in the absence of immunization. We assessed the role of serum Igs in maintaining specific B-cell subset homeostasis at steady state. Using mice genetically deficient in secreted IgM only (secretory µ chain-deficient), in switched Igs and hypermutated IgM (activation-induced cytidine deaminase-deficient), or fully agammaglobulemic (AID(-/-)µS(-/-)), we dissected the contribution of different Ig classes to 4 phenotypes associated with loss of serum Igs: 1) increased splenic B-cell numbers, mostly of the B1 and marginal zone (MZ) B-cell subtypes; 2) enlarged germinal centers (GCs) in spleen and mesenteric lymph nodes; 3) enrichment in IRF4(+)CD138(-) plasmablast-like cells; and 4) overexpression of IgM in several cell subsets. Complementation experiments based on either mixed bone marrow reconstitution of chimeras or Ig infusion, and analysis of mice raised in germ-free conditions reveal a negative feedback mechanism in which MZ and B1 cell numbers are under the control of naturally secreted Igs as the result of an intrinsic property of the immune system, whereas GC development is under indirect control of secreted Igs that limit bacterial species triggering GC reactions.


Asunto(s)
Formación de Anticuerpos/inmunología , Subgrupos de Linfocitos B/inmunología , Metagenoma , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/metabolismo , Femenino , Centro Germinal/inmunología , Inmunoglobulina M/metabolismo , Factores Reguladores del Interferón/metabolismo , Tejido Linfoide/inmunología , Ratones , Ratones Noqueados , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Bazo/inmunología , Sindecano-1/metabolismo
17.
Ann Rheum Dis ; 73(6): 1138-43, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23666932

RESUMEN

INTRODUCTION: Clinical remission is today the treatment goal for rheumatoid arthritis (RA), which requires fast and assertive therapeutic decisions for a tight control of disease activity. Few objective parameters are available to guide clinical decisions, particularly in switcher patients. We designed a preliminary algorithm introducing immunogenicity assessment in the current approach to patients with RA receiving tumour necrosis factor inhibitors (TNFi). OBJECTIVE: To evaluate the concordance between the new algorithm and current clinical practice, comparing the effectiveness of 'immunogenicity-based' versus 'empirical-based' switches in a cohort of patients with established RA receiving biologics. METHODS: EULAR therapeutic response was evaluated in 105 patients with RA (naive or switchers) over one year, through generalised estimation equation (GEE) analyses. Serum drug trough levels were assessed by ELISA and antidrug antibodies (ADAb) by Bridging ELISA. RESULTS: During follow-up, 48.6% of patients had therapeutic decisions concordant with the proposed algorithm (Group A), and 51.4% had discordant decisions (Group B). One year after the therapeutic decision, patients from Group A had a higher probability of achieving response (OR=7.91, p<0.001, 95% CI 3.27 to 19.13) and low disease activity (OR=9.77, p<0.001, 95% CI 4.69 to 20.37) than patients in Group B. CONCLUSIONS: Immunogenicity assessment might help to optimise therapeutic decisions, leading to a better control of disease activity with significantly better clinical outcomes in patients with RA receiving TNFi.


Asunto(s)
Algoritmos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos/inmunología , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adalimumab , Adulto , Anciano , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Antirreumáticos/inmunología , Artritis Reumatoide/inmunología , Productos Biológicos/inmunología , Manejo de la Enfermedad , Etanercept , Femenino , Humanos , Inmunoglobulina G/inmunología , Infliximab , Masculino , Persona de Mediana Edad , Receptores del Factor de Necrosis Tumoral/inmunología , Insuficiencia del Tratamiento , Resultado del Tratamiento
18.
Blood ; 120(23): 4552-9, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23065152

RESUMEN

Antibody-forming cells (AFCs) expressing the chemokine receptor CXCR3 are recruited to sites of inflammation where they help clear pathogens but may participate in autoimmune diseases. Here we identify a mechanism that induces CXCR3 expression by AFC and germinal center (GC) B cells. This happens when CD8 T cells are recruited into CD4 T cell-dependent B-cell responses. Ovalbumin-specific CD4 T cells (OTII) were transferred alone or with ovalbumin-specific CD8 T cells (OTI) and the response to subcutaneous alum-precipitated ovalbumin was followed in the draining lymph nodes. OTII cells alone induce T helper 2-associated class switching to IgG1, but few AFC or GC B cells express CXCR3. By contrast, OTI-derived IFN-γ induces most responding GC B cells and AFCs to express high levels of CXCR3, and diverse switching to IgG2a, IgG2b, with some IgG1. Up-regulation of CXCR3 by GC B cells and AFCs and their migration toward its ligand CXCL10 are shown to depend on B cells' intrinsic T-bet, a transcription factor downstream of the IFN-γR signaling. This model clarifies how precursors of long-lived AFCs and memory B cells acquire CXCR3 that causes their migration to inflammatory foci.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Receptores CXCR3/inmunología , Proteínas de Dominio T Box/inmunología , Vacunas/inmunología , Traslado Adoptivo , Compuestos de Alumbre , Animales , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular/inmunología , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Citometría de Flujo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunización/métodos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Regulación hacia Arriba/genética
19.
J Clin Immunol ; 33(2): 349-60, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23064977

RESUMEN

PURPOSE: Intravenous IgG (ivIg) is a therapeutic alternative for lupus erythematosus, the mechanism of which remains to be fully understood. Here we investigated whether ivIg affects two established sub-phenotypes of SLE, namely relative oligoclonality of circulating T-cells and reduced activity of CD4 + Foxp3+ regulatory T-cells (Tregs) reflected by lower CD25 surface density. METHODS: We conducted a longitudinal study of 15 lupus patients (14 with SLE and one with discoid LE) treated with ivIg in cycles of 2-6 consecutive monthly infusions. Among these 15 patients, 10 responded to ivIg therapy with clear clinical improvement. We characterized Tregs and determined TCR spectratypes of four Vß families with reported oligoclonality. Cell counts, cytometry and TCR spectratypes were obtained from peripheral blood at various time points before, during and after ivIg treatment. T-cell oligoclonality was assessed as Vß-familywise repertoire perturbation, calculated for each patient in respect to an individual reference profile averaged over all available time points. RESULTS: For 11 out of 15 patients, average Vß1/Vß2/Vß11/Vß14 repertoires were less perturbed under than outside ivIg therapy. The four exceptions with relatively increased average perturbation during ivIg therapy included three patients who failed to respond clinically to an ivIg therapy cycle. Patients' Treg CD25 surface density (cytometric MFI) was clearly reduced when compared to healthy controls, but not obviously influenced by ivIg. However, patients' average Treg CD25 MFI was found negatively correlated with both Vß11 and Vß14 perturbations measured under ivIg therapy. CONCLUSIONS: This indicates a role of active Tregs in the therapeutic effect of ivIg.


Asunto(s)
Inmunoglobulinas Intravenosas , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Factores de Transcripción Forkhead/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Estudios Longitudinales , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Adulto Joven
20.
Ann Rheum Dis ; 72(12): 1947-55, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23223420

RESUMEN

BACKGROUND: Immunogenicity of aTNFs is one of the mechanisms behind treatment failure. OBJECTIVE: To assess the effect of anti-drug antibodies (ADA) on drug response to infliximab, adalimumab and etanercept, and the effect of immunosuppression on ADA detection, in patients with Rheumatoid Arthritis, Spondyloarthritis, Psoriasis and Inflammatory Bowel Diseases. DATA SOURCES: PubMed, EMBASE, Cochrane databases, article reference lists (through August 19 2012). STUDY SELECTION: Out of 2082 studies, 17 were used in the meta-analysis (1RCT; 16 observational studies). DATA EXTRACTION: Two reviewers extracted data. Risk ratios (RR), 95% CI, using random-effect models, sensitivity analysis, meta-regressions and Egger's test were calculated. DATA SYNTHESIS: Of 865 patients, ADA against infliximab or adalimumab reduced drug response rate by 68% (RR=0.68, 95% CI=0.12 to 0.36), an effect attenuated by concomitant methotrexate (MTX): <74% MTX+: RR=0.23, 95% CI=0.15 to 0.36; ≥74% MTX+: RR=0.32, 95% CI=0.22 to 0.48. Anti-etanercept antibodies were not detected. Of 936 patients, concomitant MTX or azathioprine/mercaptopurine reduced ADA frequency by 47% (RR=0.53, 95% CI=0.42 to 0.67), particularly when ADA were assessed by RIA (RR=0.36, 95% CI=0.23 to 0.55) compared with ELISA (RR=0.63, 95% CI=0.53 to 0.74). CONCLUSIONS: ADA reduces drug response, an effect that can be attenuated by concomitant immunosuppression, which reduces ADA frequency. Drug immunogenicity should be considered for the management of patients receiving biological therapies.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adalimumab , Antiinflamatorios no Esteroideos/inmunología , Antiinflamatorios no Esteroideos/uso terapéutico , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Formación de Anticuerpos/efectos de los fármacos , Antirreumáticos/inmunología , Artritis/inmunología , Enfermedades Autoinmunes/inmunología , Medicina Basada en la Evidencia/métodos , Humanos , Inmunosupresores/farmacología , Enfermedades Inflamatorias del Intestino/inmunología , Infliximab , Metotrexato/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA