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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Immunol ; 361: 104281, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33453508

RESUMEN

Dendritic cells (DCs) mature upon an inflammatory trigger. However, an inflammatory trigger can lead to a semi-mature phenotype, allowing DCs to evoke tolerance and expedite the resolution of inflammation. This duality likely involves context-dependent modulation of inflammatory signaling. Human α1-antitrypsin (hAAT) promotes semimature DCs. We examined changes in a wide spectrum of signaling cascades in stimulated murine bone marrow-derived cells with hAAT. Upon stimulation by IL-1ß+IFNγ, hAAT-treated cells depicted an attenuated calcium flux. Disrupting PKA or NF-κB pathways revoked only some hAAT-mediated outcomes. hAAT-treated cells exhibited a distict pattern of kinase phosphorylation. hAAT-mediated increase in Treg cells in-vitro required intact inflammatory signaling pathways. Taken together, hAAT appears to require a stimulated microenvironment to promote inflammatory resolution, setting it aside from classical anti-inflammatory agents. Further studies are required to identify the specific molecules targeted by hAAT that mediate these and other outcomes.


Asunto(s)
Células Dendríticas/metabolismo , Transducción de Señal/efectos de los fármacos , alfa 1-Antitripsina/farmacología , Animales , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Calcio/metabolismo , Células Cultivadas , Tolerancia Inmunológica/inmunología , Inflamación/metabolismo , Interleucina-1/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptores CCR7/inmunología , Receptores CCR7/metabolismo , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , alfa 1-Antitripsina/metabolismo
2.
J Pharmacol Exp Ther ; 359(3): 482-490, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27821710

RESUMEN

Life-long weekly infusions of human α1-antitrypsin (hAAT) are currently administered as augmentation therapy for patients with genetic AAT deficiency (AATD). Several recent clinical trials attempt to extend hAAT therapy to conditions outside AATD, including type 1 diabetes. Since the endpoint for AATD is primarily the reduction of risk for pulmonary emphysema, the present study explores hAAT dose protocols and routes of administration in attempt to optimize hAAT therapy for islet-related injury. Islet-grafted mice were treated with hAAT (Glassia™; i.p. or s.c.) under an array of clinically relevant dosing plans. Serum hAAT and immunocyte cell membrane association were examined, as well as parameters of islet survival. Results indicate that dividing the commonly prescribed 60 mg/kg i.p. dose to three 20 mg/kg injections is superior in affording islet graft survival; in addition, a short dynamic descending dose protocol (240→120→60→60 mg/kg i.p.) is comparable in outcomes to indefinite 60 mg/kg injections. While hAAT pharmacokinetics after i.p. administration in mice resembles exogenous hAAT treatment in humans, s.c. administration better imitated the physiological progressive rise of hAAT during acute phase responses; nonetheless, only the 60 mg/kg dose depicted an advantage using the s.c. route. Taken together, this study provides a platform for extrapolating an islet-relevant clinical protocol from animal models that use hAAT to protect islets. In addition, the study places emphasis on outcome-oriented analyses of drug efficacy, particularly important when considering that hAAT is presently at an era of drug-repurposing towards an extended list of clinical indications outside genetic AATD.

3.
Proc Natl Acad Sci U S A ; 110(37): 15007-12, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23975926

RESUMEN

The rationale of α1-antitrypsin (AAT) augmentation therapy to treat progressive emphysema in AAT-deficient patients is based on inhibition of neutrophil elastase; however, the benefit of this treatment remains unclear. Here we show that clinical grade AAT (with elastase inhibitory activity) and a recombinant form of AAT (rAAT) without anti-elastase activity reduces lung inflammatory responses to LPS in elastase-deficient mice. WT and elastase-deficient mice treated with either native AAT or rAAT exhibited significant reductions in infiltrating neutrophils (23% and 68%), lavage fluid levels of TNF-α (70% and 80%), and the neutrophil chemokine KC (CXCL1) (64% and 90%), respectively. Lung parenchyma TNF-α, DNA damage-inducible transcript 3 and X-box binding protein-1 mRNA levels were reduced in both mouse strains treated with AAT; significantly lower levels of these genes, as well as IL-1ß gene expression, were observed in lungs of AAT-deficient patients treated with AAT therapy compared with untreated patients. In vitro, LPS-induced cytokines from WT and elastase-deficient mouse neutrophils, as well as neutrophils of healthy humans, were similarly reduced by AAT or rAAT; human neutrophils adhering to endothelial cells were decreased by 60-80% (P < 0.001) with either AAT or rAAT. In mouse pancreatic islet macrophages, LPS-induced surface expression of MHC II, Toll-like receptor-2 and -4 were markedly lower (80%, P < 0.001) when exposed to either AAT or rAAT. Consistently, in vivo and in vitro, rAAT reduced inflammatory responses at concentrations 40- to 100-fold lower than native plasma-derived AAT. These data provide evidence that the anti-inflammatory and immunomodulatory properties of AAT can be independent of elastase inhibition.


Asunto(s)
Antiinflamatorios/farmacología , Factores Inmunológicos/farmacología , Elastasa de Leucocito/metabolismo , Enfisema Pulmonar/tratamiento farmacológico , alfa 1-Antitripsina/farmacología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Adulto , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/deficiencia , Lipopolisacáridos/toxicidad , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Enfisema Pulmonar/genética , Enfisema Pulmonar/inmunología , Proteínas Recombinantes/farmacología , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , Deficiencia de alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/inmunología
4.
J Infect Dis ; 211(9): 1489-98, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25389308

RESUMEN

BACKGROUND: Severe bacterial infection can cause sepsis, multiple organ dysfunction syndrome (MODS), and death. Human α1-antitrypsin (hAAT) is an antiinflammatory, immune-modulating, and tissue-protective circulating serine-protease inhibitor, with levels that increase during acute-phase responses. It is currently being evaluated as a therapeutic agent for individuals with diabetes and graft-versus-host disease. However, the concern of opportunistic bacterial infections has yet to be addressed. Therefore, we investigated host immune cell responses during acute bacterial infections under conditions of elevated hAAT levels. METHODS: Peritonitis and sepsis models were created using wild-type mice and hAAT-transgenic mice. Bacterial loads, MODS, leukopenia, neutrophil infiltration, immune cell activation, circulating cytokine levels, and survival rates were then assessed. RESULTS: hAAT significantly reduced infection-induced leukopenia and liver, pancreas, and lung injury, and it significantly improved 24-hour survival rates. Unexpectedly, bacterial load was reduced. Levels of early proinflammatory mediators and neutrophil influx were increased by hAAT soon after infection but not during sterile peritonitis. CONCLUSIONS: hAAT reduces the bacterial burden after infection. Since hAAT does not block bacterial growth in culture, its effects might rely on host immune cell modulation. These outcomes suggest that prolonged hAAT treatment in patients without hAAT deficiency is safe. Additionally, hAAT treatment may be considered a preemptive therapeutic measure for individuals who are at risk for bacterial infections.


Asunto(s)
Peritonitis/microbiología , Sepsis/microbiología , alfa 1-Antitripsina/farmacología , Animales , Carga Bacteriana , Citocinas/metabolismo , Humanos , Inflamación , Leucopenia , Ratones , Ratones Transgénicos , Neutrófilos
5.
J Immunol ; 189(1): 146-53, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22634621

RESUMEN

Tolerogenic IL-10-positive CCR7-positive dendritic cells (DC) promote T regulatory (Treg) cell differentiation upon CCR7-dependent migration to draining lymph nodes (DLN). Indeed, in human DC deficiencies, Treg levels are low. α-1 antitrypsin (AAT) has been shown to reduce inflammatory markers, promote a semimature LPS-induced DC phenotype, facilitate Treg expansion, and protect pancreatic islets from alloimmune and autoimmune responses in mice. However, the mechanism behind these activities of AAT is poorly understood. In this study, we examine interactions among DC, CD4(+) T cells, and AAT in vitro and in vivo. IL-1ß/IFN-γ-mediated DC maturation and effect on Treg development were examined using OT-II cells and human AAT (0.5 mg/ml). CCL19/21-dependent migration of isolated DC and resident islet DC was assessed, and CCR7 surface levels were examined. Migration toward DLN was evaluated by FITC skin painting, transgenic GFP skin tissue grafting, and footpad DC injection. AAT-treated stimulated DC displayed reduced MHC class II, CD40, CD86, and IL-6, but produced more IL-10 and maintained inducible CCR7. Upon exposure of CD4(+) T cells to OVA-loaded AAT-treated DC, 2.7-fold more Foxp3(+) Treg cells were obtained. AAT-treated cells displayed enhanced chemokine-dependent migration and low surface CD40. Under AAT treatment (60 mg/kg), DLN contained twice more fluorescence after FITC skin painting and twice more donor DC after footpad injection, whereas migrating DC expressed less CD40, MHC class II, and CD86. Intracellular DC IL-10 was 2-fold higher in the AAT group. Taken together, these results suggest that inducible functional CCR7 is maintained during AAT-mediated anti-inflammatory conditions. Further studies are required to elucidate the mechanism behind the favorable tolerogenic activities of AAT.


Asunto(s)
Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Interleucina-10/biosíntesis , alfa 1-Antitripsina/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Células Dendríticas/metabolismo , Técnicas de Sustitución del Gen , Tolerancia Inmunológica/genética , Interleucina-10/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CCR7/fisiología , Linfocitos T Reguladores/inmunología
6.
Immunology ; 140(3): 362-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23829472

RESUMEN

B-lymphocyte activities are associated with allograft rejection. Interleukin-10 (IL-10) -expressing B cells, however, exhibit regulatory attributes. Human α1-antitrypsin (hAAT), a clinically available anti-inflammatory circulating glycoprotein that rises during acute-phase responses, promotes semi-mature dendritic cells and regulatory T (Treg) cells during alloimmune responses. Whether B lymphocytes are also targets of hAAT activity has yet to be determined. Here, we examine whether hAAT modulates B-cell responses. In culture, hAAT reduced the lipopolysaccharide-stimulated Ki-67(+) B-cell population, IgM release and surface CD40 levels, but elevated IL-10-producing cells 1.5-fold. In CD40 ligand-stimulated cultures, hAAT promoted a similar trend; reduction in the Ki-67(+) B-cell population and in surface expression of CD86, CD80 and MHCII. hAAT increased interferon-γ-stimulated macrophage B-cell activating factor (BAFF) secretion, and reduced BAFF-receptor levels. Draining lymph nodes of transgenic mice that express circulating hAAT (C57BL/6 background) and that received skin allografts exhibited reduced B-lymphocyte activation compared with wild-type recipients. BSA-vaccinated hAAT transgenic mice exhibited 2.9-fold lower BSA-specific IgG levels, but 2.3-fold greater IgM levels, compared with wild-type mice. Circulating Treg cells were 1.3-fold greater in transgenic hAAT mice, but lower in B-cell knockout (BKO) and chimeric hAAT-BKO mice, compared with wild-type mice. In conclusion, B cells are cellular targets of hAAT. hAAT-induced Treg cell expansion appears to be B-cell-dependent. These changes support the tolerogenic properties of hAAT during immune responses, and suggest that hAAT may be beneficial in pathologies that involve excessive B-cell responses.


Asunto(s)
Linfocitos B/efectos de los fármacos , Rechazo de Injerto/inmunología , Terapia de Inmunosupresión , Trasplante de Piel , alfa 1-Antitripsina/metabolismo , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Células Cultivadas , Femenino , Rechazo de Injerto/prevención & control , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina M/sangre , Interleucina-10/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Reguladores/inmunología , Trasplante Homólogo , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/inmunología
7.
Exp Cell Res ; 318(7): 789-99, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22305966

RESUMEN

The PI3K-AKT pathway is frequently activated in human cancers, including breast cancer, and its activation appears to be critical for tumor maintenance. Some malignant cells are dependent on activated AKT for their survival; tumors exhibiting elevated AKT activity show sensitivity to its inhibition, providing an Achilles heel for their treatment. Here we show that the PKCη isoform is a negative regulator of the AKT signaling pathway. The IGF-I induced phosphorylation on Ser473 of AKT was inhibited by the PKCη-induced expression in MCF-7 breast adenocarcinoma cancer cells. This was further confirmed in shRNA PKCη-knocked-down MCF-7 cells, demonstrating elevated phosphorylation on AKT Ser473. While PKCη exhibited negative regulation on AKT phosphorylation it did not alter the IGF-I induced ERK phosphorylation. However, it enhanced ERK phosphorylation when stimulated by PDGF. Moreover, its effects on IGF-I/AKT and PDGF/ERK pathways were in correlation with cell proliferation. We further show that both PKCη and IGF-I confer protection against UV-induced apoptosis and cell death having additive effects. Although the protective effect of IGF-I involved activation of AKT, it was not affected by PKCη expression, suggesting that PKCη acts through a different route to increase cell survival. Hence, our studies show that PKCη provides negative control on AKT pathway leading to reduced cell proliferation, and further suggest that its presence/absence in breast cancer cells will affect cell death, which could be of therapeutic value.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteína Quinasa C/metabolismo , Adenocarcinoma/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Quinasa C/genética , ARN Interferente Pequeño/metabolismo , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Rayos Ultravioleta
8.
Mol Med ; 17(9-10): 1000-11, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21670848

RESUMEN

Antiinflammatory clinical-grade, plasma-derived human α-1 antitrypsin (hAAT) protects islets from allorejection as well as from autoimmune destruction. hAAT also interferes with disease progression in experimental autoimmune encephalomyelitis (EAE) and in collagen-induced arthritis (CIA) mouse models. hAAT increases IL-1 receptor antagonist expression in human mononuclear cells and T-regulatory (Treg) cell population size in animal models. Clinical-grade hAAT contains plasma impurities, multiple hAAT isoforms and various states of inactive hAAT. We thus wished to establish islet-protective activities and effect on Treg cells of plasmid-derived circulating hAAT in whole animals. Islet function was assessed in mice that received allogeneic islet transplants after mice were given hydrodynamic tail-vein injection with pEF-hAAT, a previously described Epstein-Barr virus (EBV) plasmid construct containing the EBV nuclear antigen 1 (EBNA1) and the family of repeat EBNA1 binding site components (designated "EF") alongside the hAAT gene. Sera collected from hAAT-expressing mice were added to lipopolysaccharide (LPS)-stimulated macrophages to assess macrophage responsiveness. Also, maturation of peritoneal cells from hAAT-expressing mice was evaluated. hAAT-expressing mice accepted islet allografts (n = 11), whereas phosphate-buffered saline-injected animals (n = 11), as well as mice treated with truncated-hAAT-plasmid (n = 6) and untreated animals (n = 20) rapidly rejected islet allografts. In hAAT-expressing animals, local Treg cells were abundant at graft sites, and the IL-1 receptor antagonist was elevated in grafts and circulation. Sera from hAAT-expressing mice, but not control mice, inhibited macrophage responses. Finally, peritoneal cells from hAAT-expressing mice exhibited a semimature phenotype. We conclude that plasmid-derived circulating hAAT protects islet allografts from acute rejection, and human plasma impurities are unrelated to islet protection. Future studies may use this in vivo approach to examine the structure-function characteristics of the protective activities of AAT by manipulation of the hAAT plasmid.


Asunto(s)
Rechazo de Injerto/metabolismo , Inflamación/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Linfocitos T Reguladores/metabolismo , alfa 1-Antitripsina/metabolismo , Animales , Western Blotting , Línea Celular , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Rechazo de Injerto/genética , Rechazo de Injerto/prevención & control , Humanos , Sueros Inmunes/inmunología , Sueros Inmunes/farmacología , Inflamación/genética , Inflamación/prevención & control , Lipopolisacáridos/farmacología , Recuento de Linfocitos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Trasplante Homólogo , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/inmunología
9.
Biochem Biophys Res Commun ; 412(2): 313-7, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21820409

RESUMEN

The nuclear factor κB (NF-κB) family of transcription factors participates in the regulation of genes involved in innate- and adaptive-immune responses, cell death and inflammation. The involvement of the Protein kinase C (PKC) family in the regulation of NF-κB in inflammation and immune-related signaling has been extensively studied. However, not much is known on the role of PKC in NF-κB regulation in response to DNA damage. Here we demonstrate for the first time that PKC-eta (PKCη) regulates NF-κB upstream signaling by activating the IκB kinase (IKK) and the degradation of IκB. Furthermore, PKCη enhances the nuclear translocation and transactivation of NF-κB under non-stressed conditions and in response to the anticancer drug camptothecin. We and others have previously shown that PKCη confers protection against DNA damage-induced apoptosis. Our present study suggests that PKCη is involved in NF-κB signaling leading to drug resistance.


Asunto(s)
Daño del ADN , Resistencia a Antineoplásicos , FN-kappa B/agonistas , Proteína Quinasa C/metabolismo , Transporte Activo de Núcleo Celular , Antineoplásicos Fitogénicos/farmacología , Camptotecina/farmacología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Humanos , FN-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo
10.
Metab Brain Dis ; 26(2): 107-13, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21437674

RESUMEN

Alpha-1-antitrypsin (AAT) is the primary circulating serine protease inhibitor, and is known to exert potent anti-inflammatory effects and to inhibit the progression of several autoimmune diseases. In this study, transgenic mice that over-express surfactant-driven human (h)AAT on the C57BL/6 background were evaluated for resistance to MOG-35-55 peptide-induced experimental autoimmune encephalomyelitis (EAE), compared to WT C57BL/6 control mice. According to the results, sustained levels of circulating hAAT profoundly inhibited induction of clinical signs, inflammatory lesions and demyelination observed in WT mice with EAE, concomitant with enhanced levels of CD4+FoxP3+ Treg cells, reduced secretion of MOG peptide-induced pro-inflammatory cytokines, IL-17, IL-1ß & IL-6, diminished expression of caspase-1 and enhanced expression of CCR6. These results implicate hAAT as a potent immunoregulatory agent worthy of further investigation as a potential therapy in human autoimmune diseases including multiple sclerosis.


Asunto(s)
Caspasas/metabolismo , Encefalomielitis Autoinmune Experimental , Glicoproteínas/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T Reguladores/inmunología , alfa 1-Antitripsina/inmunología , Animales , Apoptosis , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Factores de Transcripción Forkhead , Glicoproteínas/administración & dosificación , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-17/análisis , Interleucina-17/metabolismo , Interleucina-1beta/análisis , Interleucina-1beta/metabolismo , Interleucina-6/análisis , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/administración & dosificación , Receptores CCR6/análisis , Receptores CCR6/metabolismo , Médula Espinal/metabolismo , Bazo/metabolismo , Linfocitos T Reguladores/metabolismo , alfa 1-Antitripsina/metabolismo
11.
Cell Signal ; 18(8): 1127-39, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16242915

RESUMEN

Protein kinase C (PKC) represents a family of serin/threonine kinases, playing a central role in the regulation of cell growth, differentiation and transformation. These enzymes differ in their primary structure, biochemical properties, tissue distribution and subcellular localization. The specific cellular functions of PKC isoforms are largely controlled by their localization. PKCeta, a member of the novel subfamily, is expressed predominantly in epithelial tissues. However, not much is known with respect to its mechanism of activation and regulation. Our recent studies suggest its role in cell cycle control. Here we show that PKCeta is localized at the Golgi apparatus, ER and the nuclear envelope. Furthermore, using GFP-fusion proteins of the different functional domains of PKCeta we deciphered the specific structural domains of the protein responsible for its apparent localization. We show that the cysteine-rich repeat C1b is responsible for its Golgi localization, while for its presence at the ER/nuclear envelope the pseudosubstrate containing fragment coupled to the C1 domain is required. In response to short-term activation by PMA we show translocation of PKCeta to the plasma membrane and the nuclear envelope. We demonstrate that the C1b is sufficient for its translocation to the plasma membrane. Interestingly, accumulation of PKCeta at the nuclear envelope also occurred in response to serum-starvation. It should be noted that interaction of PKCeta with the cyclin E/Cdk2 complex at the perinuclear region was recently reported by us in response to serum-starvation. Thus, our studies demonstrate translocation of PKCeta to the nuclear envelope, and suggest that the spatial regulation of PKCeta could be important for its cellular functions including effects on cell cycle control and involvement in tumor promotion.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Membrana Nuclear/metabolismo , Proteína Quinasa C/química , Proteína Quinasa C/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Humanos , Transporte de Proteínas/efectos de los fármacos , Factores de Tiempo
12.
J Matern Fetal Neonatal Med ; 28(5): 500-3, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24834800

RESUMEN

OBJECTIVE: To examine possible correlation between α1-antitrypsin (AAT) levels and activity in patients with and without obesity, after excluding complications such as gestational diabetes mellitus (GDM), during pregnancy. STUDY DESIGN: A prospective case-control study was conducted. AAT levels were determined by standard human AAT ELISA according to the manufacturer's instructions. Elastase inhibition was determined by kinetic assay according to manufacturer recommendations. Assays were performed in duplicates and repeated twice for each sample in separate sessions. Patients with diabetes mellitus were excluded from the study. The Mann-Whitney U-test was performed in order to determine statistical differences between the groups, and AAT concentration and activity. RESULTS: During the study period, 43 patients were recruited: 21 with isolated obesity and 22 non-obese parturients (control group). According to ELISA, AAT concentrations were mildly lower in obese women compared with non-obese women (8.31 ± 0.28 mg/ml versus 9.5 ± 0.37 mg/ml, p = 0.0155). However, the elastase inhibitory capacity was markedly lower in obese versus non-obese parturients (mean 27.33 ± 2.08 % versus 43.73 ± 3.1%, p < 0.001). CONCLUSIONS: Isolated obesity in pregnancy is associated with lower activity of AAT. These findings correlate with the reduced concentration and activity of AAT found in patients with GDM. Accordingly, it might suggest an inflammatory axis shared by obesity and the development of insulin resistance.


Asunto(s)
Obesidad/epidemiología , Complicaciones del Embarazo/epidemiología , alfa 1-Antitripsina/sangre , Adulto , Estudios de Casos y Controles , Regulación hacia Abajo , Activación Enzimática , Femenino , Humanos , Obesidad/sangre , Elastasa Pancreática/antagonistas & inhibidores , Elastasa Pancreática/metabolismo , Embarazo , Complicaciones del Embarazo/sangre , alfa 1-Antitripsina/metabolismo
13.
Cell Mol Immunol ; 11(4): 377-86, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25000533

RESUMEN

Although islet transplantation for individuals with type 1 diabetes has been shown to yield superior blood glucose control, it remains inadequate for long-term control. This is partly due to islet injuries and stresses that can lead to beta cell loss. Inhibition of excess IL-1ß activity might minimize islet injuries, thus preserving function. The IL-1 receptor antagonist (IL-1Ra), an endogenous inhibitor of IL-1ß, protects islets from cytokine-induced necrosis and apoptosis. Therefore, an imbalance between IL-1ß and IL-1Ra might influence the courses of allogeneic and autoimmune responses to islets. Our group previously demonstrated that the circulating serine-protease inhibitor human alpha-1-antitrypsin (hAAT), the levels of which increase in circulation during acute-phase immune responses, exhibits anti-inflammatory and islet-protective properties, as well as immunomodulatory activity. In the present study, we sought to determine whether the pancreatic islet allograft-protective activity of hAAT was mediated by IL-1Ra induction. Our results demonstrated that hAAT led to a 2.04-fold increase in IL-1Ra expression in stimulated macrophages and that hAAT-pre-treated islet grafts exhibited a 4.851-fold increase in IL-1Ra transcript levels, which were associated with a moderate inflammatory profile. Unexpectedly, islets that were isolated from IL-1Ra-knockout mice and pre-treated with hAAT before grafting into wild-type mice yielded an increase in intragraft IL-1Ra expression that was presumably derived from infiltrating host cells, albeit in the absence of hAAT treatment of the host. Indeed, hAAT-pre-treated islets generated hAAT-free conditioned medium that could induce IL-1Ra production in cultured macrophages. Finally, we demonstrated that hAAT promoted a distinct phosphorylation and nuclear translocation pattern for p65, a key transcription factor required for IL-1Ra expression.


Asunto(s)
Rechazo de Injerto/prevención & control , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Trasplante de Islotes Pancreáticos , Macrófagos/efectos de los fármacos , Complicaciones Posoperatorias/prevención & control , Factor de Transcripción ReIA/metabolismo , alfa 1-Antitripsina/administración & dosificación , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Células Cultivadas , Rechazo de Injerto/etiología , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/efectos de los fármacos , Factor de Transcripción ReIA/genética , Regulación hacia Arriba/efectos de los fármacos
14.
PLoS One ; 8(5): e63625, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717456

RESUMEN

Clinical pancreatic islet transplantation is under evaluation for the treatment of autoimmune diabetes, yet several limitations preclude widespread use. For example, there is a critical shortage of human pancreas donors. Xenotransplantation may solve this problem, yet it evokes a rigorous immune response which can lead to graft rejection. Alpha-1-antitrypsin (AAT), a clinically available and safe circulating anti-inflammatory and tissue protective glycoprotein, facilitates islet alloimmune-tolerance and protects from inflammation in several models. Here, we examine whether human AAT (hAAT), alone or in combination with clinically relevant approaches, achieves long-term islet xenograft survival. Rat-to-mouse islet transplantation was examined in the following groups: untreated (n = 6), hAAT (n = 6, 60-240 mg/kg every 3 days from day -10), low-dose co-stimulation blockade (anti-CD154/LFA-1) and single-dose anti-CD4/CD8 (n = 5-7), either as mono- or combination therapies. Islet grafting was accompanied by blood glucose follow-up. In addition, skin xenografting was performed in order to depict responses that occur in draining lymph nodes. According to our results hAAT monotherapy and hAAT/anti-CD154/LFA-1 combined therapy, did not delay rejection day (11-24 days untreated vs. 10-22 day treated). However, host and donor intragraft inflammatory gene expression was diminished by hAAT therapy in both setups. Single dose T-cell depletion using anti-CD4/CD8 depleting antibodies, which provided 14-15 days of reduced circulating T-cells, significantly delayed rejection day (28-52 days) but did not achieve graft acceptance. In contrast, in combination with hAAT, the group displayed significantly extended rejection days and a high rate of graft acceptance (59, 61, >90, >90, >90). In examination of graft explants, marginal mononuclear-cell infiltration containing regulatory T-cells predominated surviving xenografts. We suggest that temporal T-cell depletion, as in the clinically practiced anti-thymocyte-globulin therapy, combined with hAAT, may promote islet xenograft acceptance. Further studies are required to elucidate the mechanism behind the observed synergy, as well as the applicability of the approach for pig-to-human islet xenotransplantation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia de Injerto/inmunología , Xenoinjertos/inmunología , Islotes Pancreáticos/inmunología , alfa 1-Antitripsina/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Expresión Génica/genética , Expresión Génica/inmunología , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Supervivencia de Injerto/genética , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Trasplante Heterólogo/métodos , alfa 1-Antitripsina/genética
15.
Cell Transplant ; 22(11): 2119-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23050776

RESUMEN

Pancreatic islets are a highly vascularized entity, and their transplantation into diabetic individuals requires optimal revascularization. In addition, ß-cells in islets are extremely sensitive to inflammation. α-1-Antitrypsin (AAT), a circulating serine-protease inhibitor that is available for clinical use as an affinity-purified human product, has been shown to protect islets from graft failure in mouse transplantation models and to achieve readily vascularized islet grafts. AAT is known to induce vascular endothelial growth factor (VEGF) expression and release, as well as protect from proteolytic cleavage of VEGF by elastase, promote viability of endothelial cells, and enhance migration of myocytes. Our aim was to examine whether AAT enhances vasculogenesis toward islet grafts. We employed Matrigel-islet plugs as means to introduce islets in an explantable isolated compartment and examined vessel formation, vessel maturation, and inflammatory profile of explants 9 days after implantation. Also, we examined primary epithelial cell grafts that were prepared from lungs of mice that are transgenic for human AAT. In addition, aortic ring sprouting assay was performed, and HUVEC tube formation assays were studied in the presence of AAT. Our findings indicate that islet grafts exhibit mature vessels in the presence of AAT, as demonstrated by morphology, as well as expression of endothelial CD31, smooth muscle actin (SMA), and von Willebrand factor (vWF). Epithelial cells that express human AAT achieved a similar positive outcome. Aortic ring sprouting was enhanced in AAT-treated cultures and also in cultures that contained primary epithelial cells from human AAT transgenic animals in the absence of added AAT. According to the tube formation assay, HUVECs exhibited superior responses in the presence of AAT. We conclude that vasculogenesis toward islet grafts is enhanced in the presence of AAT. Together with the remarkable safety profile of AAT, the study supports its use in the relevant clinical setups.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/citología , alfa 1-Antitripsina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Células Cultivadas , Colágeno/química , Diabetes Mellitus Experimental/cirugía , Combinación de Medicamentos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Células Endoteliales de la Vena Umbilical Humana , Humanos , Islotes Pancreáticos/irrigación sanguínea , Laminina/química , Pulmón/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica , Proteoglicanos/química , Transcriptoma , Trasplante Homólogo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , alfa 1-Antitripsina/genética , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
16.
Front Immunol ; 4: 320, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24191154

RESUMEN

The extracellular form of the abundant heat-shock protein, gp96, is involved in human autoimmune pathologies. In patients with type 1 diabetes, circulating gp96 is found to be elevated, and is bound to the acute-phase protein, α1-antitrypsin (AAT). The two molecules also engage intracellularly during the physiological folding of AAT. AAT therapy promotes pancreatic islet survival in both transplantation and autoimmune diabetes models, and several clinical trials are currently examining AAT therapy for individuals with type 1 diabetes. However, its mechanism of action is yet unknown. Here, we examine whether the protective activity of AAT is related to binding of extracellular gp96. Primary mouse islets, macrophages, and dendritic cells were added recombinant gp96 in the presence of clinical-grade human AAT (hAAT, Glassia™, Kamada Ltd., Israel). Islet function was evaluated by insulin release. The effect of hAAT on IL-1ß/IFNγ-induced gp96 cell-surface levels was also evaluated. In vivo, skin transplantation was performed for examination of robust immune responses, and systemic inflammation was induced by cecal puncture. Endogenous gp96 was inhibited by gp96-inhibitory peptide (gp96i, Compugen Ltd., Israel) in an allogeneic islet transplantation model. Our findings indicate that hAAT binds to gp96 and diminishes gp96-induced inflammatory responses; e.g., hAAT-treated gp96-stimulated islets released less pro-inflammatory cytokines (IL-1ß by 6.16-fold and TNFα by 2.69-fold) and regained gp96-disrupted insulin release. hAAT reduced cell activation during both skin transplantation and systemic inflammation, as well as lowered inducible surface levels of gp96 on immune cells. Finally, inhibition of gp96 significantly improved immediate islet graft function. These results suggest that hAAT is a regulator of gp96-mediated inflammatory responses, an increasingly appreciated endogenous damage response with relevance to human pathologies that are exacerbated by tissue injury.

17.
J Matern Fetal Neonatal Med ; 26(18): 1782-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23650930

RESUMEN

OBJECTIVE: Alpha-1 antitrypsin (AAT), a circulating anti-inflammatory molecule, rises four- to sixfold during acute phase responses and during pregnancy. AAT deficiency is linked with various pregnancy complications. The aim of this study is to determine plasma concentrations and activity of AAT and serum cytokine levels in blood samples from women undergoing spontaneous abortions as compared with elective abortions. METHODS: A prospective case-control study consisted of patients with sporadic abortions (n = 15), recurrent spontaneous abortions (n = 14) and healthy pregnancies going through elective terminations (n = 11). Circulating AAT and cytokine levels were determined before dilatation and curettage. RESULTS: AAT levels were lower in both recurrent and sporadic spontaneous abortion groups compared with healthy pregnancies (1.421 ± 0.08, 1.569 ± 0.14 and 3.224 ± 0.45 mg/ml, respectively, p < 0.001). Reduced AAT levels correlated with elevated proinflammatory cytokines. CONCLUSIONS: AAT levels in patients with either sporadic or recurrent spontaneous abortions were lower than normal pregnancies, and were associated with an inflammatory profile. Future studies should examine larger cohort groups, effects of earlier time-points and the influence of antithrombotic therapy in such patients who are diagnosed with relatively low levels of circulating AAT, in an effort to improve pregnancy outcomes.


Asunto(s)
Aborto Espontáneo/sangre , Aborto Espontáneo/epidemiología , Deficiencia de alfa 1-Antitripsina/sangre , Deficiencia de alfa 1-Antitripsina/epidemiología , alfa 1-Antitripsina/sangre , Aborto Inducido/estadística & datos numéricos , Adulto , Estudios de Casos y Controles , Citocinas/sangre , Procedimientos Quirúrgicos Electivos/estadística & datos numéricos , Femenino , Humanos , Mediadores de Inflamación/sangre , Embarazo , Resultado del Embarazo/epidemiología , Inhibidores de Serina Proteinasa/sangre , Deficiencia de alfa 1-Antitripsina/complicaciones
18.
J Matern Fetal Neonatal Med ; 25(12): 2667-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22746289

RESUMEN

OBJECTIVE: α-1 antitrypsin (AAT) is an anti-protease, anti-inflammatory and tissue-protective molecule. Normal circulating levels are <3.5 mg/dl and rise during pregnancy. Although AAT deficiency is associated with several pregnancy and placental disorders, little is known regarding AAT levels and preeclampsia. Since unopposed inflammation might contribute to preeclampsia, we studied whether preeclampsia is associated with lower than normal levels and activity of AAT. METHODS: In a prospective case-control study, we compared maternal serum AAT activity and levels between patients with severe preeclampsia (n = 23) and without preeclampsia (n = 18). RESULTS: AAT levels were 1.91 ± 0.08-fold lower in the preeclampsia group compared to healthy group (3.854 ± 0.26 vs. 7.397 ± 0.34 mg/ml; p < 0.001), and correlated with protease inhibitory capacity (46.56 ± 2.08% vs. 67.08 ± 1.74%; p < 0.001). CONCLUSIONS: Our findings show association between lower AAT levels and severe preeclampsia during pregnancy. Further studies are required to identify the mechanism behind the association, and the possibility of safe AAT augmentation for individuals with insufficient circulating AAT.


Asunto(s)
Preeclampsia/sangre , alfa 1-Antitripsina/sangre , Adulto , Estudios de Casos y Controles , Cesárea/estadística & datos numéricos , Regulación hacia Abajo , Femenino , Humanos , Concentración Osmolar , Preeclampsia/epidemiología , Embarazo , Resultado del Embarazo/epidemiología , Índice de Severidad de la Enfermedad , Adulto Joven , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/sangre , Deficiencia de alfa 1-Antitripsina/complicaciones , Deficiencia de alfa 1-Antitripsina/epidemiología
19.
J Matern Fetal Neonatal Med ; 25(7): 934-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21843112

RESUMEN

OBJECTIVE: α1-antitrypsin (AAT) is protective of tissue damage induced by enzymes of inflammatory cell source. Inflammatory cells are involved in preterm labor, preterm premature rupture of membrane (PPROM) and term premature rupture of membrane (PROM). The purpose of this research was to examine whether plasma concentration and activity of AAT differ between these manifestations. METHODS: In a prospective case control study, blood samples were assayed for AAT concentration and activity in 71 individuals. AAT concentration and activity were measured by standard methods. RESULTS: No significant differences were found between AAT levels (p = 0.497) and activity (p = 0.879) in preterm and term labor. AAT levels and activity in PPROM and PROM were not significantly different as well (p = 0.748 and p = 0.880, respectively). While 69 out of 71 patients displayed normal circulating levels of AAT, 2 PPROM patients out of 15 had abnormally low, previously undiagnosed,AAT concentrations, and had subsequently developed complications that were absent in the other groups. CONCLUSIONS: No statistically significant differences were demonstrated in the levels of AAT between patients with preterm and term labor, nor between preterm and term PROM. Yet, unexpectedly, patients that had marked AAT deficiency belonged exclusively to the PPROM group.


Asunto(s)
Rotura Prematura de Membranas Fetales/sangre , Trabajo de Parto Prematuro/sangre , alfa 1-Antitripsina/sangre , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Embarazo , Estudios Prospectivos , Adulto Joven
20.
Diabetes ; 59(9): 2188-97, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20547979

RESUMEN

OBJECTIVE: Cellular stress and proinflammatory cytokines induce phosphorylation of insulin receptor substrate (IRS) proteins at Ser sites that inhibit insulin and IGF-1 signaling. Here, we examined the role of Ser phosphorylation of IRS-2 in mediating the inhibitory effects of proinflammatory cytokines and cellular stress on beta-cell function. RESEARCH DESIGN AND METHODS: Five potential inhibitory Ser sites located proximally to the P-Tyr binding domain of IRS-2 were mutated to Ala. These IRS-2 mutants, denoted IRS-2(5A), and their wild-type controls (IRS-2(WT)) were introduced into adenoviral constructs that were infected into Min6 cells or into cultured murine islets. RESULTS: When expressed in cultured mouse islets, IRS-2(5A) was better than IRS-2(WT) in protecting beta-cells from apoptosis induced by a combination of IL-1beta, IFN-gamma, TNF-alpha, and Fas ligand. Cytokine-treated islets expressing IRS2(5A) secreted significantly more insulin in response to glucose than did islets expressing IRS-2(WT). This could be attributed to the higher transcription of Pdx1 in cytokine-treated islets that expressed IRS-2(5A). Accordingly, transplantation of 200 islets expressing IRS2(5A) into STZ-induced diabetic mice restored their ability to respond to a glucose load similar to naïve mice. In contrast, mice transplanted with islets expressing IRS2(WT) maintained sustained hyperglycemia 3 days after transplantation. CONCLUSIONS: Elimination of a physiological negative feedback control mechanism along the insulin-signaling pathway that involves Ser/Thr phosphorylation of IRS-2 affords protection against the adverse effects of proinflammatory cytokines and improves beta-cell function under stress. Genetic approaches that promote IRS2(5A) expression in pancreatic beta-cells, therefore, could be considered a rational treatment against beta-cell failure after islet transplantation.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/fisiología , Células Secretoras de Insulina/fisiología , Insulina/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Glucemia/metabolismo , Células CHO , Caspasas/metabolismo , Cricetinae , Cricetulus , Citocinas/farmacología , Diabetes Mellitus Experimental/cirugía , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Proteínas de Homeodominio/genética , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Serina/fisiología , Transducción de Señal , Transactivadores/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA