Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 134(3)2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33328325

RESUMEN

Macrophage migration inhibitory factor (MIF) and its homologue D-dopachrome tautomerase (D-DT) are widely expressed pro-inflammatory cytokines with chemokine-like functions that coordinate a wide spectrum of biological activities, such as migration. Here, we biotin-tagged intracellular MIF/D-DT in vivo to identify important cytosolic interactors and found a plethora of actin cytoskeleton-associated proteins. Although the receptor complex between CD74 and CD44 (CD74/CD44) is essential for signalling transduction in fibroblasts via extracellular MIF/D-DT, our interactome data suggested direct effects. We, thus, investigated whether MIF/D-DT can modulate cell migration independently of CD74/CD44. To distinguish between receptor- and non-receptor-mediated motility, we used fibroblasts that are either deficient or that express CD74/CD44 proteins, and treated them with recombinant MIF/D-DT. Interestingly, only MIF could stimulate chemokinesis in the presence or absence of CD74/CD44. The pro-migratory effects of MIF depended on lipid raft/caveolae-mediated but not clathrin-mediated endocytosis, on its tautomerase activity and, probably, on its thiol protein oxidoreductase activity. As MIF treatment restrained actin polymerisation in vitro, our findings establish a new intracellular role for MIF/D-DT in driving cell motility through modulation of the actin cytoskeleton.


Asunto(s)
Movimiento Celular , Factores Inhibidores de la Migración de Macrófagos , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Células COS , Membrana Celular , Chlorocebus aethiops , Fibroblastos , Células HEK293 , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Receptores de Hialuranos , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Células 3T3 NIH , Transducción de Señal
2.
J Cell Mol Med ; 23(6): 3867-3877, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30968541

RESUMEN

Macrophage migration inhibitory factor (MIF) is pleiotropic cytokine that has multiple effects in many inflammatory and immune diseases. This study reveals a potential role of MIF in acute kidney injury (AKI) in patients and in kidney ischemic reperfusion injury (IRI) mouse model in MIF wild-type (WT) and MIF knockout (KO) mice. Clinically, plasma and urinary MIF levels were largely elevated at the onset of AKI, declined to normal levels when AKI was resolved and correlated tightly with serum creatinine independent of disease causes. Experimentally, MIF levels in plasma and urine were rapidly elevated after IRI-AKI and associated with the elevation of serum creatinine and the severity of tubular necrosis, which were suppressed in MIF KO mice. It was possible that MIF may mediate AKI via CD74/TLR4-NF-κB signalling as mice lacking MIF were protected from AKI by largely suppressing CD74/TLR-4-NF-κB associated renal inflammation, including the expression of MCP-1, TNF-α, IL-1ß, IL-6, iNOS, CXCL15(IL-8 in human) and infiltration of macrophages, neutrophil, and T cells. In conclusion, our study suggests that MIF may be pathogenic in AKI and levels of plasma and urinary MIF may correlate with the progression and regression of AKI.


Asunto(s)
Lesión Renal Aguda/sangre , Oxidorreductasas Intramoleculares/sangre , Riñón/patología , Factores Inhibidores de la Migración de Macrófagos/sangre , Daño por Reperfusión/sangre , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/orina , Adulto , Anciano , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Quimiocina CCL2/metabolismo , Creatinina/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/orina , Riñón/inmunología , Riñón/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/orina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , FN-kappa B/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/orina , Receptor Toll-Like 4/metabolismo
3.
FASEB J ; 32(8): 4107-4120, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29490169

RESUMEN

Uropathogenic Escherichia coli (UPEC) is the most common cause of urinary tract infections. In this study, UPEC strains harboring hemolysin A (HlyA) did not induce programmed cell death pathways by the activation of caspases. Instead, the UPEC pore-forming toxin HlyA triggered an increase in mitochondrial Ca2+ levels and manipulated mitochondrial dynamics by causing fragmentation of the mitochondrial network. Alterations in mitochondrial dynamics resulted in severe impairment of mitochondrial functions by loss of membrane potential, increase in reactive oxygen species production, and ATP depletion. Moreover, HlyA caused disruption of plasma membrane integrity that was accompanied by extracellular release of the danger-associated molecules high-mobility group box 1 (HMGB1) and histone 3 (H3). Our results indicate that UPEC induced programmed cell necrosis by irreversibly impairing mitochondrial function. This finding suggests a strategy devised by UPEC at the onset of infection to escape early innate immune response and silently propagate inside host cells.-Lu, Y., Rafiq, A., Zhang, Z., Aslani, F., Fijak, M., Lei, T., Wang, M., Kumar, S., Klug, J., Bergmann, M., Chakraborty, T., Meinhardt, A., Bhushan, S. Uropathogenic Escherichia coli virulence factor hemolysin A causes programmed cell necrosis by altering mitochondrial dynamics.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas Hemolisinas/metabolismo , Mitocondrias/metabolismo , Mitocondrias/fisiología , Necrosis/metabolismo , Factores de Virulencia/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Muerte Celular/fisiología , Membrana Celular/metabolismo , Proteína HMGB1/metabolismo , Histonas/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Necrosis/fisiopatología , Especies Reactivas de Oxígeno/metabolismo
4.
Mol Ther ; 26(10): 2523-2532, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30077612

RESUMEN

Macrophage migration inhibitory factor (MIF) is elevated in patients with acute kidney injury (AKI) and is suggested as a potential predictor for renal replacement therapy in AKI. In this study, we found that MIF also plays a pathogenic role and is a therapeutic target for AKI. In a cisplatin-induced AKI mouse model, elevated plasma MIF correlated with increased serum creatinine and the severity of renal inflammation and tubular necrosis, whereas deletion of MIF protected the kidney from cisplatin-induced AKI by largely improving renal functional and histological injury, and suppressing renal inflammation including upregulation of cytokines such as interleukin (IL)-1ß, tumor necrosis factor-alpha (TNF-α), IL-6, inducible nitric oxide synthase (iNOS), MCP-1, IL-8, and infiltration of macrophages, neutrophils, and T cells. We next developed a novel therapeutic strategy for AKI by blocking the endogenous MIF with an MIF inhibitor, ribosomal protein S19 (RPS19). Similar to the MIF-knockout mice, treatment with RPS19, but not the mutant RPS19, suppressed cisplatin-induced AKI. Mechanistically, we found that both genetic knockout and pharmacological inhibition of MIF protected against AKI by inactivating the CD74-nuclear factor κB (NF-κB) signaling. In conclusion, MIF is pathogenic in cisplatin-induced AKI. Targeting MIF with an MIF inhibitor RPS19 could be a promising therapeutic potential for AKI.


Asunto(s)
Lesión Renal Aguda/terapia , Inflamación/terapia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Proteínas Ribosómicas/administración & dosificación , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Apoptosis/efectos de los fármacos , Cisplatino/efectos adversos , Terapia Genética , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Riñón/efectos de los fármacos , Riñón/patología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Ratones , Ratones Noqueados , FN-kappa B/genética , Necrosis , Proteínas Ribosómicas/genética , Transducción de Señal/efectos de los fármacos
5.
Mol Hum Reprod ; 23(6): 370-380, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28379541

RESUMEN

STUDY QUESTION: What is the underlying mechanism of Sertoli cell (SC) resistance to cell death? SUMMARY ANSWER: High expression of prosurvival B-cell lymphoma-2 (BCL2) proteins and inhibition of apoptosis and autophagy prolongs SC survival upon exposure to stress stimuli. WHAT IS KNOWN ALREADY: In human and in experimental models of orchitis, tolerogenic SC survive stress conditions, while germ cells undergo massive apoptosis. In general, non-dividing highly differentiated cells tend to resist stress conditions for a longer time by favoring activation of prosurvival mechanisms and inhibition of cell death pathways. STUDY DESIGN, SIZE, DURATION: In this cross sectional study, conditions stimulating apoptosis and autophagy were used to induce cell death in primary rat SC. Primary rat peritubular cells (PTC) and immortalized rat 93RS2 SC were used as controls. Each cell isolation was counted as one experiment (n = 1), and each experiment was repeated three to six times. PARTICIPANTS/MATERIALS, SETTING, METHODS: Testis biopsy samples from infertile or subfertile patients and testis samples from rats with experimental autoimmune orchitis were used for immunohistological analysis. Primary SC were isolated from 19-day-old male Wistar rats. To maintain cell purity, cells were cultured in serum-free medium for apoptosis experiments and in medium supplemented with 1% serum for autophagy analyses. To induce apoptosis, cells were stimulated with staurosporine, borrelidin, cisplatin and etoposide for 4 or 24 h. Caspase three activation was examined by immunoblotting and enzymatic activity assay. Mitochondrial membrane potential was measured using tetramethylrhodamine methyl ester followed by flow cytometric analysis. Cytochrome c release was monitored by immunofluorescence. Cell viability was determined using the methylthiazole tetrazolium assay. To monitor autophagy flux, cells were deprived of nutrients using Hank's balanced salt solution for 1, 2 and 3 h. Formation of autophagosomes was analyzed by using immunoblotting, immunofluorescence labeling and ultrastructural analyses. Relative mRNA levels of genes involved in the regulation of apoptosis and autophagy were evaluated. Extracellular high mobility group box protein one was measured as a marker of necrosis using ELISA. MAIN RESULTS AND THE ROLE OF CHANCE: SC survive the inflammatory conditions in vivo in human testis and in experimental autoimmune orchitis. Treatment with apoptosis inducing chemotherapeutics did not cause caspase three activation in isolated rat SC. Moreover, mitochondrial membrane potential and mitochondrial localization of cytochrome c were not changed by treatment with staurosporine, suggesting a premitochondrial blockade of apoptosis in SC. Expression levels of prosurvival BCL2 family members were significantly higher in SC compared to PTC at both mRNA and protein levels. Furthermore, after nutrient starvation, autophagy signaling was initiated in SC as observed by decreased levels of phosphorylated UNC- 51-like kinase -1 (ULK1). However, levels of light chain 3 II (LC3 II) and sequestosome1 (SQSTM1) remained unchanged, indicating blockade of the autophagy flux. Lysosomal activity was intact in SC as shown by accumulation of LC3 II following administration of lysosomal protease inhibitors, indicating that inhibition of autophagy flux occurs at a preceding stage. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: In this study, we have used primary SC from prepubertal rats. Caution should be taken when translating our results to adult animals, where crosstalk with other testicular cells and hormonal factors may also play a role in regulating survival of SC. WIDER IMPLICATIONS OF THE FINDINGS: Our results suggest that inhibition of autophagy and apoptosis following exposure to extrinsic stress stimuli promotes SC survival, and is a possible mechanism to explain the robustness of SC in response to stress. Cell death resistance in SC is crucial for the recovery of spermatogenesis after chemotherapy treatment in cancer patients. Additionally, understanding the molecular mechanisms of SC survival unravels valuable target proteins, such as BCL2, that may be manipulated therapeutically to control cell viability depending on the context of the disease. STUDY FUNDING AND COMPETING INTEREST(S): This study was funded by the Deutsche Forschungsgemeinschaft (DFG) Grant BH93/1-1, and by the International Research Training Group between Justus Liebig University of Giessen and Monash University, Melbourne (GRK 1871/1) funded by the DFG and Monash University. The support of the Medical Faculty of Justus-Liebig University of Giessen is gratefully acknowledged. The authors declare no conflict of interest.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Infertilidad Masculina/genética , Orquitis/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Células de Sertoli/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Autofagia/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Supervivencia Celular/genética , Cisplatino/farmacología , Estudios Transversales , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Etopósido/farmacología , Alcoholes Grasos/farmacología , Regulación del Desarrollo de la Expresión Génica , Humanos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Orquitis/inmunología , Orquitis/patología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Células de Sertoli/efectos de los fármacos , Células de Sertoli/patología , Espermatogénesis/genética , Estaurosporina/farmacología
6.
Histochem Cell Biol ; 142(4): 421-32, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24692005

RESUMEN

Estrogen signaling is considered to play an important role in spermatogenesis, spermiogenesis and male fertility. Estrogens can act via the two nuclear estrogen receptors ESR1 (ERα) and ESR2 (ERß) or via the intracellular G-protein-coupled estrogen receptor 1 (GPER, formerly GPR30). Several reports on the localization and expression of all three receptors in the human testis have been published but are controversial particularly in case of ERα. Contrary to previous studies, we decided therefore to evaluate expression of all three receptors in the testis by a number of different methods and in comparison with MCF-7 cells. Using qPCR, we could show that mRNA expression of ERα is considerably lower and expression of ERß and GPER much higher in the testis than in MCF-7 cells. RT-PCR after laser-assisted microdissection of tubular and interstitial compartments from normal and Sertoli cell only syndrome testes plus in situ hybridization and immunohistochemical analyses of the same samples demonstrated that there is very low expression of ERα in germ cells and in single interstitial cells, very high expression of ERß in germ cells and Sertoli cells and high expression of GPER in interstitial cells and less in Sertoli cells.


Asunto(s)
Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/análisis , Receptor beta de Estrógeno/genética , Receptores de Estrógenos/análisis , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/genética , Testículo/metabolismo , Adulto , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Persona de Mediana Edad , ARN Mensajero/análisis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/química , Testículo/citología
7.
Hum Reprod ; 29(11): 2382-92, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25205753

RESUMEN

STUDY QUESTION: Is there a non-invasive biomarker for the diagnosis of testicular inflammatory lesions? SUMMARY ANSWER: In sera from infertile azoospermic patients with histologically confirmed low-grade testicular inflammation, significantly elevated titers of autoantibodies against disulfide isomerase family A, member 3 (ER-60) were found. WHAT IS KNOWN ALREADY: Infection and inflammation of the genital tract are supposed to be responsible for up to 15% of cases among infertile males. However, specific seminal or serological markers are not available to assess subacute or chronic inflammatory conditions in the testis. STUDY DESIGN, SIZE, DURATION: This study consisted of the identification of autoantibodies for testicular antigens in sera of patients with low-grade testicular inflammation, validation of candidates, development of an ELISA for the most promising target antigen and measurement of autoantibodies titers in healthy normozoospermic men (n = 20); male blood donors (n = 14); men with impaired semen quality without (n = 14) or with (n = 26) symptoms of genital tract infection/inflammation; azoospermic men with histologically confirmed testicular inflammatory lesions (n = 16); men after pharmacotherapy of genital tract infection/inflammation (n = 15) and men with acute epididymo-orchitis (n = 30). PARTICIPANTS/MATERIALS, SETTING, METHODS: Proteins in lysates of normal testicular tissue were separated by high-resolution 2D gel electrophoresis and probed with sera of 13 patients with histologically confirmed chronic testicular inflammation. There were 14 proteins that immunoreacted with a majority of these sera and could be identified by mass spectrometry. Of these 14 proteins, disulfide isomerase family A, member 3 (ER-60), transferrin and chaperonin containing TCP1 complex, subunit 5 (epsilon) (CCT5) were considered as specific. Since ER-60 reacted with 92% of patient sera, an ER-60-autoantibody ELISA was developed. MAIN RESULTS AND THE ROLE OF CHANCE: The newly established ELISA detected significantly elevated titers of autoantibodies against ER-60 in the sera from infertile men with histologically confirmed chronic testicular inflammation (median 8.6; P < 0.01) compared with the control groups. Moreover, elevated levels of anti-ER-60 titers were detected in patients suffering from acute epididymo-orchitis (median 3.3; P < 0.05) as compared with healthy normozoospermic men (median 2.13; P < 0.001), male blood donors with unknown fertility status (median 2.72; P < 0.01), patients with impaired semen quality but no infection/inflammation (median 2.59; P < 0.001) and patients with symptoms of genital tract infections and/or inflammation (median 2.18; P < 0.001). Significantly lower levels of anti-ER-60 antibodies were measured in sera from patients after application of anti-inflammatory pharmacotherapy (median 1.9; P < 0.01) compared with those with histologically confirmed chronic testicular inflammation. The cut-off value of the assay was set to 6.6 U/ml based on a calculated sensitivity of 100% and a specificity of 81.2%. LIMITATIONS, REASONS FOR CAUTION: The results obtained in this study showed statistically significant elevated titers of ER-60 antibodies in sera from patients with histologically confirmed testicular inflammatory lesions and from a few patients with acute epididymo-orchitis. However, the number of serum samples tested was limited. Severe testicular damage seen in azoospermic patients could represent a bias towards ER-60 reactivity, while the assay does not allow for different etiologies of the lesions to be distinguished. Due to ethical reasons, the prevalence of testicular inflammatory lesions among controls and non-azoospermic men cannot be studied at the histological level. WIDER IMPLICATIONS OF THE FINDINGS: Measurement of ER-60 autoantibody titers in serum could be a novel non-invasive marker for the diagnosis of asymptomatic testicular inflammation causing male fertility disturbances. STUDY FUNDING/COMPETING INTERESTS: This study was supported by a grant of the Deutsche Forschungsgemeinschaft (ME 1323/4-4) and the Translational Science Fund (Wirtschafts-und Strukturbank Hessen-WI Bank). M.F., A.P., W.W., H.-C.S. and A.M. are supported by the LOEWE focus group 'MIBIE' (Male infertility during infection and inflammation). The ER-60 ELISA is protected by a patent to the Justus-Liebig-University of Giessen with A.M. and M.F. as inventors (patent no. DE 10 2008 053 503). T.Z. as employee of the DRG Company was responsible for the ELISA development.


Asunto(s)
Autoanticuerpos/análisis , Infertilidad Masculina/diagnóstico , Inflamación/diagnóstico , Proteína Disulfuro Isomerasas/inmunología , Testículo/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Azoospermia/diagnóstico , Azoospermia/inmunología , Azoospermia/patología , Biomarcadores/análisis , Humanos , Infertilidad Masculina/inmunología , Infertilidad Masculina/patología , Inflamación/inmunología , Inflamación/patología , Masculino , Persona de Mediana Edad , Análisis de Semen , Adulto Joven
8.
Clin Sci (Lond) ; 124(10): 627-37, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23252627

RESUMEN

RPS19 (ribosomal protein S19), a component of the 40S small ribosomal subunit, has recently been identified to bind the pro-inflammatory cytokine macrophage MIF (migration inhibitory factor). In vitro experiments identify RPS19 as the first endogenous MIF inhibitor by blocking the binding of MIF to its receptor CD74 and MIF functions on monocyte adherence to endothelial cells. In the present study, we sought to establish whether recombinant RPS19 can exert anti-inflammatory effects in a mouse model of anti-GBM (glomerular basement membrane) GN (glomerulonephritis) in which MIF is known to play an important role. Accelerated anti-GBM GN was induced in C57BL/6J mice by immunization with sheep IgG followed 5 days later by administration of sheep anti-mouse GBM serum. Groups of eight mice were treated once daily by intraperitoneal injection with 6 mg of RPS19/kg of body weight or an irrelevant control protein (human secretoglobin 2A1), or received no treatment, from day 0 until being killed on day 10. Mice that received control or no treatment developed severe crescentic anti-GBM disease on day 10 with increased serum creatinine, declined creatinine clearance and increased proteinuria. These changes were associated with up-regulation of MIF and its receptor CD74 activation of ERK (extracellular-signal-regulated kinase) and NF-κB (nuclear factor κB) signalling, prominent macrophage and T-cell infiltration, as well as up-regulation of Th1 [T-bet and IFNγ (interferon γ)] and Th17 [STAT3 (signal transducer and activator of transcription 3) and IL (interleukin)-17A] as well as IL-1ß and TNFα (tumour necrosis factor α). In contrast, RPS19 treatment largely prevented the development of glomerular crescents and glomerular necrosis, and prevented renal dysfunction and proteinuria (all P<0.001). Of note, RPS19 blocked up-regulation of MIF and CD74 and inactivated ERK and NF-κB signalling, thereby inhibiting macrophage and T-cell infiltration, Th1 and Th17 responses and up-regulation of pro-inflammatory cytokines (all P<0.01). These results demonstrate that RPS19 is a potent anti-inflammatory agent, which appears to work primarily by inhibiting MIF signalling.


Asunto(s)
Enfermedad por Anticuerpos Antimembrana Basal Glomerular/fisiopatología , Antiinflamatorios/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Proteínas Ribosómicas/uso terapéutico , Animales , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/tratamiento farmacológico , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/prevención & control , Antígenos de Diferenciación de Linfocitos B/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Membrana Basal Glomerular/inmunología , Antígenos de Histocompatibilidad Clase II/biosíntesis , Interferón gamma , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Ovinos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
9.
J Immunol ; 186(9): 5162-72, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21441459

RESUMEN

Despite the immune-privileged status of the male genital tract, infection and inflammation of the male genital tract are important etiological factors in male infertility. A common observation in clinical and experimental orchitis as well as in systemic infection and inflammation are decreased levels of testosterone. Emerging data point to an immunosuppressive role of testosterone. In our study, we substituted testosterone levels in experimental autoimmune orchitis (EAO) in rat by s.c. testosterone implants. EAO development was reduced to 17% when animals were treated with low-dose testosterone implants (3 cm long, EAO+T3) and to 33% when rats were supplied with high-dose testosterone implants (24 cm, EAO+T24) compared with 80% of animals developing disease in the EAO control group. In the testis, testosterone replacement in EAO animals prevented the accumulation of macrophages and significantly reduced the number of CD4(+) T cells with a strong concomitant increase in the number of regulatory T cells (CD4(+)CD25(+)Foxp3(+)) compared with EAO control. In vitro testosterone treatment of naive T cells led to an expansion of the regulatory T cell subset with suppressive activity and ameliorated MCP-1-stimulated chemotaxis of T lymphocytes in a Transwell assay. Moreover, expression of proinflammatory mediators such as MCP-1, TNF-α, and IL-6 in the testis and secretion of Th1 cytokines such as IFN-γ and IL-2 by mononuclear cells isolated from testicular draining lymph nodes were decreased in the EAO+T3 and EAO+T24 groups. Thus, our study shows an immunomodulatory and protective effect of testosterone substitution in the pathogenesis of EAO and suggests androgens as a new factor in the differentiation of regulatory T cells.


Asunto(s)
Andrógenos/inmunología , Orquitis/inmunología , Linfocitos T Reguladores/inmunología , Testosterona/inmunología , Andrógenos/farmacología , Animales , Separación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Microscopía Fluorescente , Orquitis/tratamiento farmacológico , Ratas , Ratas Endogámicas WKY , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testosterona/farmacología
10.
J Biol Chem ; 284(50): 34944-53, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19826004

RESUMEN

Ubiquitinated proteins can alternatively be delivered directly to the proteasome or via p97/VCP (valosin-containing protein). Whereas the proteasome degrades ubiquitinated proteins, the homohexameric ATPase p97/VCP seems to control the ubiquitination status of recruited substrates. The COP9 signalosome (CSN) is also involved in the ubiquitin/proteasome system (UPS) as exemplified by regulating the neddylation of ubiquitin E3 ligases. Here, we show that p97/VCP colocalizes and directly interacts with subunit 5 of the CSN (CSN5) in vivo and is associated with the entire CSN complex in an ATP-dependent manner. Furthermore, we provide evidence that the CSN and in particular the isopeptidase activity of its subunit CSN5 as well as the associated deubiquitinase USP15 are required for proper processing of polyubiquitinated substrates bound to p97/VCP. Moreover, we show that in addition to NEDD8, CSN5 binds to oligoubiquitin chains in vitro. Therefore, CSN and p97/VCP could form an ATP-dependent complex that resembles the 19 S proteasome regulatory particle and serves as a key mediator between ubiquitination and degradation pathways.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complejos Multiproteicos/metabolismo , Péptido Hidrolasas/metabolismo , Adenosina Trifosfatasas/genética , Secuencia de Aminoácidos , Animales , Complejo del Señalosoma COP9 , Proteínas de Ciclo Celular/genética , Humanos , Ratones , Datos de Secuencia Molecular , Complejos Multiproteicos/genética , Células 3T3 NIH , Péptido Hidrolasas/genética , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ubiquitinación , Proteína que Contiene Valosina
11.
Clin Chem ; 55(5): 946-54, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19264855

RESUMEN

BACKGROUND: Serum samples from patients with autoimmune connective tissue diseases that show a finely speckled antinuclear antibody (ANA) on indirect immune-fluorescence often have antibodies against unknown nuclear target antigens. To search for such autoantigens we applied a proteomic approach using sera from patients with a high ANA titer (>or=640) and finely speckled fluorescence but in whom no antibodies to extractable nuclear antigens (ENA) could be identified. METHODS: Using an immunoproteomics approach we identified heterogeneous nuclear ribonucleoprotein H1 (hnRNP H1) as a novel nuclear target of autoantibody response. RESULTS: Recombinant rat hnRNP H1 reacted in Western blot analyses with 48% of 93 sera from patients with primary Sjögren syndrome and with 5.2% of 153 sera from patients with other connective tissue diseases (diseased controls). For comparison, the diagnostic sensitivity and specificity of anti-Sjögren syndrome A (SSA) antibodies for primary Sjögren syndrome in the same patient cohort were 88.2% and 76.3%, respectively. Interestingly, 5 of 11 primary Sjögren syndrome patients with no anti-SSA or anti-SSB antibodies had anti-hnRNP H1 antibodies. Anti-hnRNP H1 antibodies were preabsorbed by hnRNP H1, as demonstrated by indirect immunofluorescence. In an evaluation of the presence of anti-hnRNP H1 antibodies in 188 consecutive samples submitted to the clinical laboratory with positive ANA (titer >or=160), anti-hnRNP H1 antibodies were found in 3 of 7 (2 primary and 5 secondary) Sjögren syndrome patients and in 8.3% of the diseased controls. CONCLUSIONS: HnRNP H1 is a newly discovered autoantigen that could become an additional diagnostic marker.


Asunto(s)
Autoanticuerpos/inmunología , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/inmunología , Síndrome de Sjögren/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Autoanticuerpos/sangre , Western Blotting , Estudios de Cohortes , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/sangre , Humanos , Masculino , Persona de Mediana Edad , Curva ROC , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Síndrome de Sjögren/sangre , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
12.
Sci Rep ; 8(1): 3741, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29487346

RESUMEN

Galectin-1 (Gal-1) is a pleiotropic lectin involved in the modulation of immune responses. Using a model of rat experimental autoimmune orchitis (EAO), we investigated the role of Gal-1 in testicular inflammation. EAO is characterized by leukocytic infiltrates in the interstitium, damage of spermatogenesis and production of inflammatory mediators like TNFα and MCP1 causing infertility. In normal rat testis Gal-1 was mainly expressed in Sertoli cells and germ cells. In the inflamed testis, Gal-1 expression was significantly downregulated most likely due to germ cell loss. Analyses of lectin binding and expression of glucosaminyl- and sialyltransferases indicated that the glycan composition on the cell surface of Sertoli and peritubular cells becomes less favourable for Gal-1 binding under inflammatory conditions. In primary Sertoli cells Gal-1 expression was found to be upregulated after TNFα challenge. Pretreatment with Gal-1 synergistically and specifically enhanced TNFα-induced expression of MCP1, IL-1α, IL-6 and TNFα in Sertoli cells. Combined stimulation of Sertoli cells with Gal-1 and TNFα enhanced the phosphorylation of MAP kinases as compared to TNFα or Gal-1 alone. Taken together, our data show that Gal-1 modulates inflammatory responses in Sertoli cells by enhancing the pro-inflammatory activity of TNFα via stimulation of MAPK signalling.


Asunto(s)
Galectina 1/metabolismo , Sistema de Señalización de MAP Quinasas , Orquitis/etiología , Orquitis/metabolismo , Células de Sertoli/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Galectina 1/genética , Expresión Génica , Células Germinativas/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Masculino , Orquitis/patología , ARN Mensajero/genética , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Mol Endocrinol ; 19(12): 2964-78, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16020486

RESUMEN

Human secretoglobin (SCGB) 2A1 (or lipophilin C, lacryglobin, mammaglobin B) is a small protein of unknown function that forms heterodimers with secretoglobin 1D1 (lipophilin A) in tears and is expressed in the prostate. Here we show that SCGB 2A1 is under androgen control in the androgen-responsive prostatic cell line LNCaP and can be induced more than 20-fold by dihydrotestosterone. Only 6 h after androgen treatment, a strong DNase I-hypersensitive site is induced in the proximal promoter within chromatin. Within the boundaries of this DNase I-hypersensitive site a minimal 32-bp peculiar dimeric inverted repeat variant GC box (dim-IR-GA box) was found to confer androgen but not glucocorticoid responsiveness in gene transfer experiments. Mutations of both GA boxes that abolish binding of Sp1 and Sp3 also abrogate the androgen response. In an EMSA the DNA binding domain of the androgen receptor (AR) was not able to bind directly to the dim-IR-GA box. However, AR is functionally required for the hormone response because induction can be inhibited with the nonsteroidal antagonist bicalutamide. Chromatin immunoprecipitation experiments demonstrated that AR is recruited to the proximal promoter 10 min after androgen treatment. Therefore we propose that SCGB 2A1 represents a new class of androgen target genes that are purely under indirect AR control mediated by DNA-bound Sp factors.


Asunto(s)
Andrógenos/metabolismo , Proteínas Portadoras/genética , Regulación de la Expresión Génica , Próstata/metabolismo , Elementos de Respuesta/genética , Factores de Transcripción Sp/metabolismo , Antagonistas de Andrógenos/farmacología , Andrógenos/farmacología , Anilidas/farmacología , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Cromatina/efectos de los fármacos , Dihidrotestosterona/farmacología , Humanos , Inmunoprecipitación , Masculino , Mamoglobina B , Datos de Secuencia Molecular , Mutación , Proteínas de la Mielina , Factores de Transcripción NFI/metabolismo , Nitrilos , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína , Proteolípidos , Secuencias Repetitivas de Ácidos Nucleicos/genética , Secretoglobinas , Compuestos de Tosilo , Uteroglobina
14.
J Vis Exp ; (108): e53389, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26890157

RESUMEN

The testis, and in particular the male gamete, challenges the immune system in a unique way because differentiated sperm first appear at the time of puberty - more than ten years after the establishment of systemic immune tolerance. Spermatogenic cells express a number of proteins that may be seen as non-self by the immune system. The testis must then be able to establish tolerance to these neo-antigens on the one hand but still be able to protect itself from infections and tumor development on the other hand. Therefore the testis is one of a few immune privileged sites in the body that tolerate foreign antigens without evoking a detrimental inflammatory immune response. Sertoli cells play a key role for the maintenance of this immune privileged environment of the testis and also prolong survival of cotransplanted cells in a foreign environment. Therefore primary Sertoli cells are an important tool for studying the immune privilege of the testis that cannot be easily replaced by established cell lines or other cellular models. Here we present a detailed and comprehensive protocol for the isolation of Sertoli cells - and peritubular cells if desired - from rat testes within a single day.


Asunto(s)
Tolerancia Inmunológica , Inmunidad Celular , Células de Sertoli/citología , Testículo/citología , Animales , Línea Celular , Masculino , Ratas , Células de Sertoli/inmunología , Testículo/inmunología
15.
J Androl ; 26(3): 379-86, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15867006

RESUMEN

Leydig cells have been implicated in several inflammation-related responses of the testis. Specifically, these cells produce the proinflammatory cytokines interleukin-1 (IL-1) and IL-6, stimulate macrophage recruitment, and promote interstitial fluid formation. In addition, the immunoregulatory cytokines macrophage migration inhibitory factor (MIF), transforming growth factor-beta1 (TGFbeta1), and interferon-gamma (IFNgamma) are constitutively expressed by testicular cells, including the Leydig cells. In the present study, the contribution of the Leydig cell to testicular inflammatory responses was examined in adult male rats treated with the Leydig cell-specific toxin, ethane dimethane sulfonate (EDS). Intratesticular testosterone levels were modulated by subcutaneous testosterone implants. After 10 days, animals received an injection of lipopolysaccharide (LPS) to induce an inflammatory response, or saline alone, and were killed 3 hours later. Both depletion of Leydig cells by EDS and LPS treatment caused a decrease in collected testicular interstitial fluid to about 35% of control levels, but the effects were not additive. Maintenance of intratesticular testosterone reversed the interstitial fluid decline following EDS treatment and partially prevented the LPS-induced effect. MIF, TGFbeta1, and IFNgamma were expressed in both the normal and inflamed testis at similar levels. In contrast, EDS treatment caused a significant decline in expression of all 3 cytokines, which was prevented by the testosterone implants. These data indicate that 1) expression of TGFbeta1, MIF, and IFNgamma in the testis is not dependent on the presence of intact Leydig cells but is under direct testosterone control and 2) the decline in testicular interstitial fluid during inflammation involves the Leydig cells, acting via both androgens and nonandrogenic secretions. These data provide further support for a significant role for the Leydig cell in modulating the testicular response to inflammation.


Asunto(s)
Citocinas/biosíntesis , Células Intersticiales del Testículo/fisiología , Orquitis/inmunología , Animales , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/fisiología , Expresión Génica/fisiología , Interferón gamma/biosíntesis , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/inmunología , Lipopolisacáridos , Factores Inhibidores de la Migración de Macrófagos/biosíntesis , Masculino , Mesilatos/farmacología , Orquitis/metabolismo , Orquitis/patología , Ratas , Ratas Sprague-Dawley , Testosterona/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta1
16.
Reprod Fertil Dev ; 17(4): 435-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15899155

RESUMEN

Cytokines have direct effects on testicular cell functions and a number of cytokines are produced constitutively within the testis, even in the absence of immune-activation events. There is clear evidence that cytokines play a dual role as important regulatory factors in the normal function of the testis, as well as in testicular inflammation. The pro-inflammatory cytokine macrophage migration inhibitory factor (MIF) is expressed locally in the testis and has direct effects on peritubular cells, which, in turn, produce anti-inflammatory mediators, including transforming growth factor (TGF)-(2)2. In the present study, we investigated the function of MIF by examining its effect on the secretion of TGF-(2)2 in peritubular cells. Expression of TGF-(2)2 mRNA was shown by reverse transcription-polymerase chain reaction in peritubular cells isolated from 19-day-old rat testis. The addition of recombinant MIF to cultured peritubular cells resulted in a dose-dependent decrease in TGF-(2)2 secretion up to 52% of control levels after 48 h, which was significant for all doses investigated (10-100 ng mL(-1) MIF). Inhibition of TGF-(2)2 secretion was sustained for 72 h for the highest dose of MIF used (100 ng mL(-1)). No effect of MIF was observed on TGF-(2)2 mRNA expression levels, as shown by real-time polymerase chain reaction. These results suggest that the pro-inflammatory cytokine MIF can shift the cytokine balance from the immunosuppressive state towards an inflammatory reaction, potentially through the inhibition of TGF-(2)2 secretion by peritubular cells.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/metabolismo , Túbulos Seminíferos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/farmacología , Masculino , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Túbulos Seminíferos/citología , Túbulos Seminíferos/efectos de los fármacos , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta2
17.
Mol Biol Cell ; 26(15): 2845-57, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26063731

RESUMEN

CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells are able to inhibit proliferation and cytokine production in effector T-cells and play a major role in immune responses and prevention of autoimmune disease. A master regulator of Treg cell development and function is the transcription factor Foxp3. Several cytokines, such as TGF-ß and IL-2, are known to regulate Foxp3 expression as well as methylation of the Foxp3 locus. We demonstrated previously that testosterone treatment induces a strong increase in the Treg cell population both in vivo and in vitro. Therefore we sought to investigate the direct effect of androgens on expression and regulation of Foxp3. We show a significant androgen-dependent increase of Foxp3 expression in human T-cells from women in the ovulatory phase of the menstrual cycle but not from men and identify a functional androgen response element within the Foxp3 locus. Binding of androgen receptor leads to changes in the acetylation status of histone H4, whereas methylation of defined CpG regions in the Foxp3 gene is unaffected. Our results provide novel evidence for a modulatory role of androgens in the differentiation of Treg cells.


Asunto(s)
Factores de Transcripción Forkhead/sangre , Receptores Androgénicos/sangre , Linfocitos T Reguladores/metabolismo , Adulto , Diferenciación Celular/fisiología , Femenino , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Receptores Androgénicos/genética , Linfocitos T Reguladores/citología
19.
PLoS One ; 6(12): e28452, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22164293

RESUMEN

Infertility affects one in seven couples and ascending bacterial infections of the male genitourinary tract by Escherichia coli are an important cause of male factor infertility. Thus understanding mechanisms by which immunocompetent cells such as testicular macrophages (TM) respond to infection and how bacterial pathogens manipulate defense pathways is of importance. Whole genome expression profiling of TM and peritoneal macrophages (PM) infected with uropathogenic E. coli (UPEC) revealed major differences in regulated genes. However, a multitude of genes implicated in calcium signaling pathways was a common feature which indicated a role of calcium-dependent nuclear factor of activated T cells (NFAT) signaling. UPEC-dependent NFAT activation was confirmed in both cultured TM and in TM in an in vivo UPEC infectious rat orchitis model. Elevated expression of NFATC2-regulated anti-inflammatory cytokines was found in TM (IL-4, IL-13) and PM (IL-3, IL-4, IL-13). NFATC2 is activated by rapid influx of calcium, an activity delineated to the pore forming toxin alpha-hemolysin by bacterial mutant analysis. Alpha-hemolysin suppressed IL-6 and TNF-α cytokine release from PM and caused differential activation of MAP kinase and AP-1 signaling pathways in TM and PM leading to reciprocal expression of key pro-inflammatory cytokines in PM (IL-1α, IL-1ß, IL-6 downregulated) and TM (IL-1ß, IL-6 upregulated). In addition, unlike PM, LPS-treated TM were refractory to NFκB activation shown by the absence of degradation of IκBα and lack of pro-inflammatory cytokine secretion (IL-6, TNF-α). Taken together, these results suggest a mechanism to the conundrum by which TM initiate immune responses to bacteria, while maintaining testicular immune privilege with its ability to tolerate neo-autoantigens expressed on developing spermatogenic cells.


Asunto(s)
Escherichia coli/metabolismo , Regulación de la Expresión Génica , Sistema Inmunológico , Infertilidad Masculina/diagnóstico , Orquitis/microbiología , Animales , Calcio/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas Hemolisinas/metabolismo , Infertilidad Masculina/microbiología , Macrófagos/citología , Masculino , Factores de Transcripción NFATC/metabolismo , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Orquitis/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Espermatogénesis , Testículo/microbiología
20.
J Biol Chem ; 284(12): 7977-85, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19155217

RESUMEN

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been implicated in the pathogenesis of inflammatory disorders such as infection, sepsis, and autoimmune disease. MIF exists preformed in cytoplasmic pools and exhibits an intrinsic tautomerase and oxidoreductase activity. MIF levels are elevated in the serum of animals and patients with infection or different inflammatory disorders. To elucidate how MIF actions are controlled, we searched for endogenous MIF-interacting proteins with the potential to interfere with key MIF functions. Using in vivo biotin-tagging and endogenous co-immunoprecipitation, the ribosomal protein S19 (RPS19) was identified as a novel MIF binding partner. Surface plasmon resonance and pulldown experiments with wild type and mutant MIF revealed a direct physical interaction of the two proteins (K(D) = 1.3 x 10(-6) m). As RPS19 is released in inflammatory lesions by apoptotic cells, we explored whether it affects MIF function and inhibits its binding to receptors CD74 and CXCR2. Low doses of RPS19 were found to strongly inhibit MIF-CD74 interaction. Furthermore, RPS19 significantly compromised CXCR2-dependent MIF-triggered adhesion of monocytes to endothelial cells under flow conditions. We, therefore, propose that RPS19 acts as an extracellular negative regulator of MIF.


Asunto(s)
Células Endoteliales/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Monocitos/metabolismo , Receptores de Interleucina-8B/metabolismo , Proteínas Ribosómicas/metabolismo , Animales , Apoptosis/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Adhesión Celular/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Mutación , Células 3T3 NIH , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Unión Proteica/genética , Ratas , Receptores de Interleucina-8B/agonistas , Receptores de Interleucina-8B/genética , Proteínas Ribosómicas/genética , Sepsis/genética , Sepsis/metabolismo , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA