Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Immunol ; 17(5): 556-64, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26974206

RESUMEN

Homeostasis of the immune system depends on the proper function of regulatory T cells (T(reg) cells). Compromised suppressive activity of T(reg) cells leads to autoimmune disease and graft rejection and promotes anti-tumor immunity. Here we report a previously unrecognized requirement for the serine-threonine phosphatase PP2A in the function of T(reg) cells. T(reg) cells exhibited high PP2A activity, and T(reg) cell-specific ablation of the PP2A complex resulted in a severe, multi-organ, lymphoproliferative autoimmune disorder. Mass spectrometry revealed that PP2A associated with components of the mTOR metabolic-checkpoint kinase pathway and suppressed the activity of the mTORC1 complex. In the absence of PP2A, T(reg) cells altered their metabolic and cytokine profile and were unable to suppress effector immune responses. Therefore, PP2A is required for the function of T(reg) cells and the prevention of autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Trastornos Linfoproliferativos/inmunología , Proteína Fosfatasa 2/inmunología , Linfocitos T Reguladores/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/genética , Autoinmunidad/inmunología , Células Cultivadas , Ceramidas/inmunología , Ceramidas/metabolismo , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Células Jurkat , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Fosforilación/inmunología , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
2.
Clin Exp Rheumatol ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38910570

RESUMEN

OBJECTIVES: The aim of these studies was to characterise the molecular effects of a tool JAK1 inhibitor on cultured primary fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA) through both total and individual cell analysis. METHODS: RA-FLS cultures from 6 (Bulk RNA-seq) or 4 (ScRNA-seq) donors were pre-treated with various concentrations (100 nM and 1µM) of ABT-317 with/without exposure to 25% SEB-conditioned PBMC medium to mimic the RA inflammatory milieu. Cells were subjected to both bulk RNA-seq (36 libraries) and single cell RNA-seq (scRNA-seq; 24 libraries) to identify biological processes impacted by CM and ABT-317 treatments. RESULTS: In our bulk RNA-seq analysis, a total of 2,605 differentially expressed genes (DEGs) were identified between CM-stimulation and unstimulated groups, while 1,122 DEGs were found between ABT-317 1µM and DMSO in CM-stimulated groups using thresholds of log2 (fold change) ≥ |0.58| and FDR ≤ 10%. Both bulk and single cell mRNA analysis of RA-FLS treated with a combination of CM and ABT-317 demonstrated the expected changes in inflammatory pathways such as interferon and IL-6 signalling. However, other non-inflammation associated pathways were also altered by ABT-317. In addition, the single cell analysis highlighted that FLS segregate into distinctive clusters upon combination CM and ABT-317 treatment, suggesting JAK inhibition can drive RA-FLS into multiple heterogenous cell populations. Interestingly, one of the unique RA-FLS clusters that emerged from the CM and ABT-317 treatment showed matrix metalloproteinase-3 (MMP3)high expression as well as several gene signatures that are not found in any other ABT-317 derived clusters. CONCLUSIONS: JAK inhibition with ABT-317 is effective at globally inhibiting CM-induced pro- and non-inflammatory pathways in FLS cultures, but also results in several distinct fibroblast populations with unique gene-associated pathways. This study advances the molecular understanding of JAK1 inhibitor effects on fibroblasts that may contribute to clinical efficacy.

3.
Int J Mol Sci ; 25(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38791146

RESUMEN

Crohn's disease (CD) is a subtype of inflammatory bowel disease (IBD) characterized by transmural disease. The concept of transmural healing (TH) has been proposed as an indicator of deep clinical remission of CD and as a predictor of favorable treatment endpoints. Understanding the pathophysiology involved in transmural disease is critical to achieving these endpoints. However, most studies have focused on the intestinal mucosa, overlooking the contribution of the intestinal wall in Crohn's disease. Multi-omics approaches have provided new avenues for exploring the pathogenesis of Crohn's disease and identifying potential biomarkers. We aimed to use transcriptomic and proteomic technologies to compare immune and mesenchymal cell profiles and pathways in the mucosal and submucosa/wall compartments to better understand chronic refractory disease elements to achieve transmural healing. The results revealed similarities and differences in gene and protein expression profiles, metabolic mechanisms, and immune and non-immune pathways between these two compartments. Additionally, the identification of protein isoforms highlights the complex molecular mechanisms underlying this disease, such as decreased RTN4 isoforms (RTN4B2 and RTN4C) in the submucosa/wall, which may be related to the dysregulation of enteric neural processes. These findings have the potential to inform the development of novel therapeutic strategies to achieve TH.


Asunto(s)
Colon , Enfermedad de Crohn , Mucosa Intestinal , Proteómica , Humanos , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Enfermedad de Crohn/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteómica/métodos , Colon/metabolismo , Colon/patología , Transcriptoma , Masculino , Femenino , Adulto , Perfilación de la Expresión Génica , Biomarcadores , Persona de Mediana Edad , Multiómica
4.
J Immunol ; 206(8): 1719-1728, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33762326

RESUMEN

Protein phosphatase 2A (PP2A) composed of a scaffold subunit, a catalytic subunit, and multiple regulatory subunits is a ubiquitously expressed serine/threonine phosphatase. We have previously shown that the PP2A catalytic subunit is increased in T cells from patients with systemic lupus erythematosus and promotes IL-17 production by enhancing the activity of Rho-associated kinase (ROCK) in T cells. However, the molecular mechanism whereby PP2A regulates ROCK activity is unknown. In this study, we show that the PP2A regulatory subunit PPP2R2A is increased in T cells from people with systemic lupus erythematosus and binds to, dephosphorylates, and activates the guanine nucleotide exchange factor GEF-H1 at Ser885, which in turn increases the levels of RhoA-GTP and the activity of ROCK in T cells. Genetic PPP2R2A deficiency in murine T cells reduced Th1 and Th17, but not regulatory T cell differentiation and mice with T cell-specific PPP2R2A deficiency displayed less autoimmunity when immunized with myelin oligodendrocyte glycoprotein peptide. Our studies indicate that PPP2R2A is the regulatory subunit that dictates the PP2A-directed enhanced Th1 and Th17 differentiation, and therefore, it represents a therapeutic target for pathologies linked to Th1 and Th17 cell expansion.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Proteína Fosfatasa 2/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Animales , Hidrolasas de Éster Carboxílico/genética , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Lupus Eritematoso Sistémico/genética , Activación de Linfocitos , Ratones , Ratones Noqueados , Proteína Fosfatasa 2/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
5.
J Immunol ; 200(9): 3077-3086, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29602774

RESUMEN

Cytotoxic function and cytokine profile of NK cells are compromised in patients with systemic lupus erythematosus (SLE). CD3ζ, an important molecule for NK cell activation, is downregulated in SLE T cells and contributes to their altered function. However, little is known about the role of CD3ζ in SLE NK cells. We studied CD3ζ levels and its contribution to cytotoxic, degranulation, and cytokine production capacity of NK cells from patients with SLE. Furthermore, we studied the human NK cell line, NKL, in which manipulation of CD3ζ levels was achieved using small interfering RNA and NK cells from Rag2 mice deficient in CD3ζ. We found reduced CD3ζ expression in NK cells from SLE patients independent of disease activity. Downregulation of CD3ζ expression in NK cells is mediated, at least in part, by Caspase 3, the activity of which is higher in NK cells from patients with SLE compared with NK cells from healthy donors. CD3ζ levels correlated inversely with natural cytotoxicity and the percentage of cells capable of producing the proinflammatory cytokines IFN-γ and TNF. In contrast, CD3ζ levels showed a direct correlation with levels of Ab-dependent cellular cytotoxicity. Experiments performed in CD3ζ-silenced NKL and CD3ζ-deficient NK cells from Rag2 mice confirmed the dependence of NK cell function on CD3ζ levels. Our results demonstrate a differential role for CD3ζ in natural cytotoxicity and Ab-dependent cellular cytotoxicity. We conclude that downregulated CD3ζ confers a proinflammatory phenotype to SLE NK cells and contributes to their altered function in patients with SLE.


Asunto(s)
Complejo CD3/inmunología , Células Asesinas Naturales/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Complejo CD3/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Activación de Linfocitos/inmunología , Ratones , Fenotipo
6.
Crit Rev Immunol ; 37(1): 39-58, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29431078

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease linked to profound defects in the function and phenotype of T lymphocytes. Here, we describe abnormal signaling pathways that have been documented in T cells from patients with SLE and discuss how they impact gene expression and immune function, in order to understand how they contribute to disease development and progression.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Linfocitos T/inmunología , Animales , Humanos , Activación de Linfocitos/inmunología , Fenotipo , Transducción de Señal/inmunología
7.
J Immunol ; 193(2): 544-54, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24935930

RESUMEN

Systemic lupus erythematosus (SLE) is a human chronic inflammatory disease generated and maintained throughout life by autoreactive T and B cells. Class I phosphoinositide 3-kinases (PI3K) are heterodimers composed of a regulatory and a catalytic subunit that catalyze phosphoinositide-3,4,5-P3 formation and regulate cell survival, migration, and division. Activity of the PI3Kδ isoform is enhanced in human SLE patient PBLs. In this study, we analyzed the effect of inhibiting PI3Kδ in MRL/lpr mice, a model of human SLE. We found that PI3Kδ inhibition ameliorated lupus progression. Treatment of these mice with a PI3Kδ inhibitor reduced the excessive numbers of CD4(+) effector/memory cells and B cells. In addition, this treatment reduced serum TNF-α levels and the number of macrophages infiltrating the kidney. Expression of inactive PI3Kδ, but not deletion of the other hematopoietic isoform PI3Kγ, reduced the ability of macrophages to cross the basement membrane, a process required to infiltrate the kidney, explaining MRL/lpr mice improvement by pharmacologic inhibition of PI3Kδ. The observations that p110δ inhibitor prolonged mouse life span, reduced disease symptoms, and showed no obvious secondary effects indicates that PI3Kδ is a promising target for SLE.


Asunto(s)
Adenosina/análogos & derivados , Riñón/efectos de los fármacos , Lupus Eritematoso Sistémico/prevención & control , Macrófagos/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinonas/farmacología , Adenosina/química , Adenosina/farmacología , Animales , Anticuerpos Antinucleares/sangre , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Inmunoglobulina G/sangre , Riñón/metabolismo , Riñón/patología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Nefritis Lúpica/prevención & control , Recuento de Linfocitos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Microscopía Confocal , Estructura Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/química , Quinazolinonas/química , Quinoxalinas/farmacología , Análisis de Supervivencia , Tiazolidinedionas/farmacología
8.
J Allergy Clin Immunol ; 133(1): 130-8.e1-2, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24290282

RESUMEN

BACKGROUND: Sublingual administration of Phleum pratense allergen immunotherapy (SLIT) tablets is a clinically efficient treatment for grass pollen-induced rhinoconjunctivitis. This immunotherapy downregulates TH2 immune responses, induces tolerogenic pathways, and increases regulatory T cells. However, associated immune response markers of allergen desensitization remain undefined. OBJECTIVE: We sought to characterize the kinetics of individual changes in the immunologic response to grass tablet SLIT. METHODS: We evaluated the systemic effects of SLIT in a longitudinal analysis of humoral and cellular immune parameters in peripheral blood samples. RESULTS: Grass tablet SLIT administration induced a 2-phase systemic humoral and cellular response. The TH2 response was initially exacerbated and detected as increased allergen-specific IgE (sIgE) and IgG4 (sIgG4) levels and an increase in IL-4-producing cells, followed by downregulation of the TH2 response with a shift toward a TH1 cytokine profile. T cells with a regulatory phenotype were also elicited. Statistical correlations between immunologic measurements for each patient throughout therapy indicated that TH2 response downregulation and reduction of the immediate SLIT-induced IgE response were associated with increased allergen-specific IgG4 synthesis early in therapy. TH2 response downregulation by month 4 correlated with increased frequency of CD4(+) T cells with a regulatory phenotype by 12 months. CONCLUSION: Changes in sIgE levels after therapy were linked to a specific IgG4 response, and production of blocking antibodies correlated with TH2 response downregulation. Reduced IL-4(+) cell frequency was linked to an increase in the frequency of CD4(+) T cells with a regulatory phenotype. Changes in sIgE levels and reduced IL-4 and blocking antibody levels could thus be used as indicators of a patient's immune response to therapy.


Asunto(s)
Phleum/inmunología , Extractos Vegetales/uso terapéutico , Rinitis Alérgica Estacional/tratamiento farmacológico , Inmunoterapia Sublingual/métodos , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th2/inmunología , Adulto , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Inmunomodulación , Interleucina-4/metabolismo , Rinitis Alérgica Estacional/inmunología , Comprimidos
9.
J Immunol ; 187(5): 2376-85, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21810603

RESUMEN

Systemic lupus erythematosus (SLE) is a human chronic inflammatory disease caused by the action of autoreactive T and B cells. Class I phosphoinositide-3-kinases (PI3K) are enzymes that trigger formation of 3-poly-phosphoinositides that induce cell survival. Enhanced PI3K activation is a frequent event in human cancer. Nonetheless, in a genetic model with enhanced activation of class I(A) PI3K in T cells, mice show a greater tumor index but die of a lupus-like disease. In this study, we studied the potential PI3K involvement in human SLE. The PI3K pathway was frequently activated in SLE patient PBMC and T cells (∼70% of cases), more markedly in active disease phases. We examined the mechanism for PI3K pathway activation and found enhanced activation of PI3Kδ in SLE peripheral blood T cells. The magnitude of PI3K pathway activation in patients paralleled activated/memory T cell accumulation. We examined potential tolerance mechanisms affected by increased PI3K activity; SLE patients showed reduced activation-induced cell death of activated/memory T cells. Moreover, the defective activation-induced cell death in SLE T cells was corrected after reduction of PI3Kδ activity, suggesting that PI3Kδ contributes to induction of enhanced SLE memory T cell survival. These observations point to PI3Kδ as a target of clinical interest for SLE.


Asunto(s)
Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Linfocitos T/inmunología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Adulto , Apoptosis/inmunología , Western Blotting , Separación Celular , Supervivencia Celular/inmunología , Activación Enzimática/inmunología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Memoria Inmunológica , Masculino , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T/citología , Adulto Joven
10.
Mucosal Immunol ; 15(6): 1338-1349, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36372810

RESUMEN

Inflammatory bowel disease (IBD) is characterized by a dysregulated intestinal epithelial barrier leading to breach of barrier immunity. Here we identified similar protein expression changes between IBD and Citrobacter rodentium-infected FVB mice with respect to dysregulation of solute transporters as well as components critical for intestinal barrier integrity. We attribute the disease associated changes in the model to the emergence of undifferentiated intermediate intestinal epithelial cells. Prophylactic treatment with IL-22.Fc in C. rodentium-infected FVB mice reduced disease severity and rescued the mice from lethality. Multi-omics and solute analyses revealed that IL-22.Fc treatment prevented disease-associated changes including disruption of the solute transporter machinery and restored proper physiological functions of the intestine, respectively. Taken together, we established the disease relevance of the C. rodentium-induced colitis model to IBD, demonstrated the protective role of IL-22 in amelioration of epithelial dysfunction and elucidated the molecular mechanisms with IL-22's effect on intestinal epithelial cells.


Asunto(s)
Colitis , Infecciones por Enterobacteriaceae , Enfermedades Inflamatorias del Intestino , Interleucinas , Animales , Ratones , Citrobacter rodentium/fisiología , Colitis/tratamiento farmacológico , Colitis/microbiología , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/metabolismo , Intestinos , Ratones Endogámicos C57BL , Interleucinas/farmacología , Interleucina-22
11.
Cell Rep ; 30(1): 112-123.e4, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31914379

RESUMEN

Patients with systemic lupus erythematosus (SLE) suffer frequent infections that account for significant morbidity and mortality. T cell cytotoxic responses are decreased in patients with SLE, yet the responsible molecular events are largely unknown. We find an expanded CD8CD38high T cell subset in a subgroup of patients with increased rates of infections. CD8CD38high T cells from healthy subjects and patients with SLE display decreased cytotoxic capacity, degranulation, and expression of granzymes A and B and perforin. The key cytotoxicity-related transcription factors T-bet, RUNX3, and EOMES are decreased in CD8CD38high T cells. CD38 leads to increased acetylated EZH2 through inhibition of the deacetylase Sirtuin1. Acetylated EZH2 represses RUNX3 expression, whereas inhibition of EZH2 restores CD8 T cell cytotoxic responses. We propose that high levels of CD38 lead to decreased CD8 T cell-mediated cytotoxicity and increased propensity to infections in patients with SLE, a process that can be reversed pharmacologically.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Linfocitos T CD8-positivos/inmunología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Infecciones/inmunología , Lupus Eritematoso Sistémico/inmunología , NAD/metabolismo , Sirtuina 1/metabolismo , Linfocitos T Citotóxicos/inmunología , Adulto , Línea Celular , Femenino , Humanos , Lupus Eritematoso Sistémico/microbiología , Masculino , Factores de Transcripción/metabolismo
12.
Arthritis Rheumatol ; 71(1): 99-108, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30058241

RESUMEN

OBJECTIVE: Signaling lymphocytic activation molecule family member 1 (SLAMF1) homophilic interactions promote immunoglobulin production and T cell-B cell cross-talk. SLAMF1 is overexpressed on T and B cells in patients with systemic lupus erythematosus (SLE). This study was undertaken to determine the role of SLAMF1 monoclonal antibody (mAb) in modulating T cell-B cell interaction and B cell activation. METHODS: Anti-IgM-prestimulated naive or total B cells from either healthy donors or patients with SLE were cocultured with autologous T cells under CD3/CD28 stimulation, in the presence or absence of the SLAMF1 mAb. Naive B cells were stimulated with anti-IgM and CD40L in the presence of the SLAMF1 antibody. Cytokine production by CD4+ T cells and B cells was examined by flow cytometry and/or quantitative polymerase chain reaction. Plasmablast formation and T cell and B cell conjugates were assessed by flow cytometry. IgG and antinuclear antibody production was determined by enzyme-linked immunosorbent assay. RESULTS: SLAMF1 ligation in a human peripheral blood T cell-B cell culture system reduced the following in both healthy controls and patients with SLE: conjugate formation, interleukin-6 (IL-6) production by B cells, IL-21 and IL-17A production by T cells, and Ig and autoantibody production. Whereas the SLAMF1 mAb directly affected the function of isolated peripheral B cells by decreasing IL-6 and Ig production in vitro, it did not affect cytokine production by isolated T cells stimulated in vitro. CONCLUSION: The SLAMF1 antibody inhibits T cell-B cell interaction and suppresses B cell cytokine production and differentiation, thereby acting as a potential therapeutic tool in the treatment of patients with SLE.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Células Plasmáticas/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Linfocitos T/inmunología , Adulto , Linfocitos B/efectos de los fármacos , Estudios de Casos y Controles , Técnicas de Cocultivo , Femenino , Humanos , Interleucina-17/inmunología , Interleucina-6/inmunología , Interleucinas/inmunología , Linfopoyesis/efectos de los fármacos , Linfopoyesis/inmunología , Masculino , Persona de Mediana Edad , Células Plasmáticas/citología , Células Plasmáticas/efectos de los fármacos , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Células Th17/efectos de los fármacos , Células Th17/inmunología
13.
Front Immunol ; 10: 2172, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31572382

RESUMEN

Hyaluronic acid (HA), a component of the extracellular matrix, is the ligand for CD44 and has been implicated in the pathogenesis of kidney inflammation in patients with systemic lupus erythematosus (SLE), but its direct role and mechanism of action have not been studied. Here we show that administration of hymecromone (4-Methylumbelliferone, 4-MU), an HA synthesis inhibitor, to lupus-prone mice suppressed dramatically lupus-related pathology. Interestingly, 4-MU stopped the appearance of disease when administered prior to its onset and inhibited the progression of disease when administered after its appearance. Inhibition of HA synthesis in vivo reduced tissue damage and the number of intrarenal lymphoid cell infiltrates including double negative CD3+CD4-CD8- T cells which are known to be involved in the pathogenesis of SLE. Exposure of human peripheral blood mononuclear cells to HA in vitro increased the generation of CD3+CD4-CD8- T cells through a mechanism involving Rho-associated kinase. Our results signify the importance of the HA-rich tissue microenvironment in the activation of lymphocytes to cause tissue damage in SLE and suggest the consideration of inhibition of HA synthesis to treat patients.


Asunto(s)
Ácido Hialurónico/biosíntesis , Riñón/patología , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Piel/patología , Adulto , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/antagonistas & inhibidores , Himecromona/farmacología , Indicadores y Reactivos/farmacología , Riñón/efectos de los fármacos , Riñón/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/prevención & control , Masculino , Ratones Endogámicos MRL lpr , Piel/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
14.
Arthritis Rheumatol ; 71(11): 1869-1878, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31233276

RESUMEN

OBJECTIVE: Glutaminase 1 (Gls1) is the first enzyme in glutaminolysis. The selective Gls1 inhibitor bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES) suppresses Th17 development and ameliorates experimental autoimmune encephalomyelitis (EAE). The present study was undertaken to investigate whether inhibition of glutaminolysis is beneficial for the treatment of systemic lupus erythematosus (SLE), and the involved mechanisms. METHODS: MRL/lpr mice were treated with BPTES or vehicle control, and disease activity was examined. Then naive CD4+ T cells from patients with SLE were cultured under Th17-polarizing conditions with BPTES or vehicle. Furthermore, using newly generated Gls1 conditional-knockout mice, in vitro Th17 differentiation was examined, and EAE was induced in the mice. Glutaminolysis and glycolysis were measured with an extracellular flux analyzer. The expression of hypoxia-inducible factor 1α (HIF-1α) was examined by Western blotting. RESULTS: Treatment of MRL/lpr mice with BPTES improved autoimmune pathology in a Th17-dependent manner. T cells from patients with SLE treated with BPTES displayed decreased Th17 differentiation (P < 0.05). Using the conditional-knockout mice, we demonstrated that both in vitro Th17 differentiation (P < 0.05) and the development of EAE were dependent on Gls1. Gls1 inhibition reduced glycolysis and the expression of HIF-1α protein, which induces glycolysis. CONCLUSION: We demonstrated that inhibition of glutaminolysis represents a potential new treatment strategy for patients with SLE and Th17-related autoimmune diseases. Mechanistically, we have shown that inhibition of glutaminolysis affects the glycolysis pathway by reducing HIF-1α protein in Th17 cells.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/genética , Glutaminasa/antagonistas & inhibidores , Glucólisis/efectos de los fármacos , Lupus Eritematoso Sistémico/genética , Sulfuros/farmacología , Tiadiazoles/farmacología , Adulto , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Glutaminasa/genética , Glutamina/efectos de los fármacos , Glutamina/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Técnicas In Vitro , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Ratones Noqueados , Persona de Mediana Edad , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo
15.
Cancer Immunol Res ; 7(9): 1485-1496, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315913

RESUMEN

The tumor microenvironment in leukemia and solid tumors induces a shift of activated CD8+ cytotoxic T cells to an exhausted state, characterized by loss of proliferative capacity and impaired immunologic synapse formation. Efficient strategies and targets need to be identified to overcome T-cell exhaustion and further improve overall responses in the clinic. Here, we took advantage of the Eµ-TCL1 chronic lymphocytic leukemia (CLL) and B16 melanoma mouse models to assess the role of the homophilic cell-surface receptor SLAMF6 as an immune-checkpoint regulator. The transfer of SLAMF6+ Eµ-TCL1 cells into SLAMF6-/- recipients, in contrast to wild-type (WT) recipients, significantly induced expansion of a PD-1+ subpopulation among CD3+CD44+CD8+ T cells, which had impaired cytotoxic functions. Conversely, administering anti-SLAMF6 significantly reduced the leukemic burden in Eµ-TCL1 recipient WT mice concomitantly with a loss of PD-1+CD3+CD44+CD8+ T cells with significantly increased effector functions. Anti-SLAMF6 significantly reduced leukemic burden in the peritoneal cavity, a niche where antibody-dependent cellular cytotoxicity (ADCC) is impaired, possibly through activation of CD8+ T cells. Targeting of SLAMF6 affected tumor growth not only in B cell-related leukemia and lymphomas but also in nonhematopoietic tumors such as B16 melanoma, where SLAMF6 is not expressed. In vitro exhausted CD8+ T cells showed increased degranulation when anti-human SLAMF6 was added in culture. Taken together, anti-SLAMF6 both effectively corrected CD8+ T-cell dysfunction and had a direct effect on tumor progression. The outcomes of our studies suggest that targeting SLAMF6 is a potential therapeutic strategy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Inmunomodulación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Biomarcadores , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Inmunomodulación/genética , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/patología , Melanoma Experimental , Ratones , Ratones Noqueados , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Microambiente Tumoral/inmunología
16.
Nat Rev Nephrol ; 13(6): 329-343, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28287110

RESUMEN

Glomerulonephritis is traditionally considered to result from the invasion of the kidney by autoantibodies and immune complexes from the circulation or following their formation in situ, and by cells of the innate and the adaptive immune system. The inflammatory response leads to the proliferation and dysfunction of cells of the glomerulus, and invasion of the interstitial space with immune cells, resulting in tubular cell malfunction and fibrosis. T cells are critical drivers of autoimmunity and related organ damage, by supporting B-cell differentiation and antibody production or by directly promoting inflammation and cytotoxicity against kidney resident cells. T cells might become activated by autoantigens in the periphery and become polarized to secrete inflammatory cytokines before entering the kidney where they have the opportunity to expand owing to the presence of costimulatory molecules and activating cytokines. Alternatively, naive T cells could enter the kidney where they become activated after encountering autoantigen and expand locally. As not all individuals with a peripheral autoimmune response to kidney antigens develop glomerulonephritis, the contribution of local kidney factors expressed or produced by kidney cells is probably of crucial importance. Improved understanding of the biochemistry and molecular biology of T cells in patients with glomerulonephritis offers unique opportunities for the recognition of treatment targets for autoimmune kidney disease.


Asunto(s)
Autoinmunidad , Enfermedades Renales/inmunología , Linfocitos T/fisiología , Animales , Autoanticuerpos/inmunología , Citocinas/fisiología , Glomerulonefritis/inmunología , Humanos , Inmunidad Innata , Inflamación/inmunología , Lupus Eritematoso Sistémico/inmunología
17.
Trends Mol Med ; 23(7): 615-635, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28623084

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease affecting multiple organs. A complex interaction of genetics, environment, and hormones leads to immune dysregulation and breakdown of tolerance to self-antigens, resulting in autoantibody production, inflammation, and destruction of end-organs. Emerging evidence on the role of these factors has increased our knowledge of this complex disease, guiding therapeutic strategies and identifying putative biomarkers. Recent findings include the characterization of genetic/epigenetic factors linked to SLE, as well as cellular effectors. Novel observations have provided an improved understanding of the contribution of tissue-specific factors and associated damage, T and B lymphocytes, as well as innate immune cell subsets and their corresponding abnormalities. The intricate web of involved factors and pathways dictates the adoption of tailored therapeutic approaches to conquer this disease.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Linfocitos T/inmunología , Animales , Linfocitos B/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/terapia , Linfocitos T/patología
18.
Curr Opin Immunol ; 43: 32-38, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27636649

RESUMEN

Systemic Lupus Erythematosus is an autoimmune disorder caused by a complex combination of genetic, epigenetic and environmental factors. Different polymorphisms and epigenetic modifications lead to altered gene expression and function of several molecules which lead to abnormal T cell responses. Metabolic and functional alterations result in peripheral tolerance failures and biased differentiation of T cells into pro-inflammatory and B cell-helper phenotypes as well as the accumulation of disease-promoting memory T cells. Understanding these T cell alterations and their origins is necessary to develop more accurate patient classification systems and to discover new therapeutic targets.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/inmunología , Subgrupos de Linfocitos T/inmunología , Células Th17/inmunología , Animales , Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Comunicación Celular , Diferenciación Celular , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Terapia Molecular Dirigida , Transducción de Señal
19.
PLoS One ; 10(11): e0141171, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26544975

RESUMEN

BACKGROUND: T cells regulate the adaptive immune response and have altered function in autoimmunity. Systemic Lupus Erythematosus (SLE) has great diversity of presentation and treatment response. Peripheral blood component gene expression affords an efficient platform to investigate SLE immune dysfunction and help guide diagnostic biomarker development for patient stratification. METHODS: Gene expression in peripheral blood T cell samples for 14 SLE patients and 4 controls was analyzed by high depth sequencing. Unbiased clustering of genes and samples revealed novel patterns related to disease etiology. Functional annotation of these genes highlights pathways and protein domains involved in SLE manifestation. RESULTS: We found transcripts for hundreds of genes consistently altered in SLE T cell samples, for which DAVID analysis highlights induction of pathways related to mitochondria, nucleotide metabolism and DNA replication. Fewer genes had reduced mRNA expression, and these were linked to signaling, splicing and transcriptional activity. Gene signatures associated with the presence of dsDNA antibodies, low complement levels and nephritis were detected. T cell gene expression also indicates the presence of several patient subtypes, such as having only a minimal expression phenotype, male type, or severe with or without induction of genes related to membrane protein production. CONCLUSIONS: Unbiased transcriptome analysis of a peripheral blood component provides insight on autoimmune pathophysiology and patient variability. We present an open source workflow and richly annotated dataset to support investigation of T cell biology, develop biomarkers for patient stratification and perhaps help indicate a source of SLE immune dysfunction.


Asunto(s)
Biomarcadores/metabolismo , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Lupus Eritematoso Sistémico/clasificación , Lupus Eritematoso Sistémico/genética , Linfocitos T/metabolismo , Transcriptoma/genética , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Lupus Eritematoso Sistémico/patología , Masculino , Estructura Terciaria de Proteína , Linfocitos T/patología
20.
PLoS One ; 9(6): e99607, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24932813

RESUMEN

INTRODUCTION: Synovial fibroblasts (SF) undergo phenotypic changes in rheumatoid arthritis (RA) that contribute to inflammatory joint destruction. This study was undertaken to evaluate the clinical and functional significance of ectopic podoplanin (gp38) expression by RA SF. METHODS: Expression of gp38 and its CLEC2 receptor was analyzed by immunohistochemistry in synovial arthroscopic biopsies from RA patients and normal and osteoarthritic controls. Correlation between gp38 expression and RA clinicopathological variables was analyzed. In patients rebiopsied after anti-TNF-α therapy, changes in gp38 expression were determined. Platelet-SF coculture and gp38 silencing in SF were used to analyze the functional contribution of gp38 to SF migratory and invasive properties, and to SF platelet crosstalk. RESULTS: gp38 was abundantly but variably expressed in RA, and it was undetectable in normal synovial tissues. Among clinicopathologigal RA variables, significantly increased gp38 expression was only found in patients with lymphoid neogenesis (LN), and RF or ACPA autoantibodies. Cultured synovial but not dermal fibroblasts showed strong constitutive gp38 expression that was further induced by TNF-α. In RA patients, anti-TNF-α therapy significantly reduced synovial gp38 expression. In RA synovium, CLEC2 receptor expression was only observed in platelets. gp38 silencing in cultured SF did not modify their migratory and invasive properties but reduced the expression of IL-6 and IL-8 genes induced by SF-platelet interaction. CONCLUSIONS: In RA, synovial expression of gp38 is strongly associated to LN and it is reduced after anti-TNF-α therapy. Interaction between gp38 and CLEC2 platelet receptor is feasible in RA synovium in vivo and can specifically contribute to gene expression by SF.


Asunto(s)
Artritis Reumatoide/metabolismo , Plaquetas/fisiología , Fibroblastos/fisiología , Glicoproteínas de Membrana/fisiología , Membrana Sinovial/metabolismo , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Plaquetas/metabolismo , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/patología , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inflamación , Lectinas Tipo C/biosíntesis , Lectinas Tipo C/genética , Tejido Linfoide/patología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Osteoartritis/metabolismo , Osteoartritis/patología , ARN Interferente Pequeño/farmacología , Células del Estroma/patología , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA