Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Biol ; 17(9): e3000451, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31525189

RESUMEN

Nucleotide-binding, leucine-rich repeat containing X1 (NLRX1) is a mitochondria-located innate immune sensor that inhibits major pro-inflammatory pathways such as type I interferon and nuclear factor-κB signaling. We generated a novel, spontaneous, and rapidly progressing mouse model of multiple sclerosis (MS) by crossing myelin-specific T-cell receptor (TCR) transgenic mice with Nlrx1-/- mice. About half of the resulting progeny developed spontaneous experimental autoimmune encephalomyelitis (spEAE), which was associated with severe demyelination and inflammation in the central nervous system (CNS). Using lymphocyte-deficient mice and a series of adoptive transfer experiments, we demonstrate that genetic susceptibility to EAE lies within the innate immune compartment. We show that NLRX1 inhibits the subclinical stages of microglial activation and prevents the generation of neurotoxic astrocytes that induce neuronal and oligodendrocyte death in vitro. Moreover, we discovered several mutations within NLRX1 that run in MS-affected families. In summary, our findings highlight the importance of NLRX1 in controlling the early stages of CNS inflammation and preventing the onset of spontaneous autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/etiología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Adulto , Animales , Astrocitos/fisiología , Estudios de Casos y Controles , Sistema Nervioso Central/patología , Codón sin Sentido , Enfermedades Desmielinizantes , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inmunidad Innata , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Mutación Missense , Adulto Joven
2.
Cytokine ; 124: 154501, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30097285

RESUMEN

The hepatic fibrogenic response is a protective mechanism activated by hepatocyte damage and is resolved upon elimination of the cause. However, persistent injuries cause liver fibrosis (LF) to evolve into cirrhosis, which promotes the development of hepatocellular carcinoma (HCC). Development of efficient treatments for LF requires better understanding the underlying molecular pathogenic mechanisms. The loss of suppressor of cytokine signaling 1 (SOCS1) expression promotes LF and HCC in human and mice, but the underlying mechanisms remain unclear. SOCS1 is a key regulator of immune cell activation. To investigate the anti-fibrogenic functions of SOCS1 in hepatocytes and macrophages, we generated mice lacking SOCS1 in hepatocytes (Socs1fl/flAlbCre) or macrophages (Socs1fl/flLysMCre) and evaluated hepatic fibrogenic response to carbon tetrachloride (CCl4). Socs1fl/flAlbCre and Socs1fl/flLysMCre mice showed severe LF characterized by increased collagen deposition, hydroxyproline content, myofibroblast accumulation along with elevated expression of Acta2 and Col1a1 genes. CCl4 treatment triggered significant damage to hepatocytes in Socs1fl/flAlbCre mice but not in Socs1fl/flLysMCre mice. In both mice CCl4 treatment reduced the expression of Mmp2 and increased the expression of Timp1. SOCS1 deficiency in hepatocytes or macrophages did not affect Il6, Tnfa or Tgfb, but diminished Infg and augmented Pdgfb expression. Both Socs1fl/flAlbCre and Socs1fl/flLysMCre livers showed increased mononuclear cell infiltration accompanied by elevated Ccl2 expression. Our findings show that SOCS1 exerts non-redundant functions in hepatocytes and macrophages to regulate the hepatic fibrogenic response possibly through limiting hepatocyte damage and the inflammatory response of macrophages, and support the idea of exploiting SOCS1 in LF treatment.


Asunto(s)
Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Hígado/fisiopatología , Macrófagos/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Quimiocina CCL2/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Citocinas/metabolismo , Regulación de la Expresión Génica/genética , Inflamación/metabolismo , Leucocitos/metabolismo , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/enzimología , Cirrosis Hepática/patología , Linfocinas/genética , Linfocinas/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
3.
Curr Top Microbiol Immunol ; 410: 159-189, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28900678

RESUMEN

SOCS1 is a negative feedback regulator of cytokine and growth factor receptor signaling, and plays an indispensable role in attenuating interferon gamma signaling. Studies on SOCS1-deficient mice have established a crucial role for SOCS1 in regulating CD8+ T cell homeostasis. In the thymus, SOCS1 prevents thymocytes that had failed positive selection from surviving and expanding, ensures negative selection and prevents inappropriate developmental skewing toward the CD8 lineage. In the periphery, SOCS1 not only controls production of T cell stimulatory cytokines but also attenuates the sensitivity of CD8+ T cells to synergistic cytokine stimulation and antigen non-specific activation. As cytokine stimulation of CD8+ T lymphocytes increases their sensitivity to low affinity TCR ligands, SOCS1 likely contributes to peripheral T cell tolerance by putting brakes on aberrant T cell activation driven by inflammatory cytokines. In addition, SOCS1 is critical to maintain the stability of T regulatory cells and control their plasticity to become pathogenic Th17 and Th1 cells under the harmful influence of inflammatory cytokines. SOCS1 also regulates T cell activation by dendritic cells via modulating their generation, maturation, antigen presentation, costimulatory signaling, and cytokine production. The above control mechanisms of SOCS1 on T cells, T regulatory cells and dendritic cells collectively contribute to immunological tolerance and prevent autoimmune manifestation. On other hand, silencing SOCS1 in dendritic cells or CD8+ T cells stimulates efficient antitumor immunity. Thus, even though SOCS1 is not a cell surface checkpoint inhibitor, its regulatory functions on T cell responses qualify SOCS1as a "non-classical" checkpoint blocker. SOCS1 also functions as a tumor suppressor in cancer cells by regulating oncogenic signal transduction pathways. The loss of SOCS1 expression observed in many tumors may have an impact on classical checkpoint pathways. The potential to exploit SOCS1 to treat inflammatory/autoimmune diseases and elicit antitumor immunity is discussed.


Asunto(s)
Autoinmunidad , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Animales , Humanos , Ratones , Ratones Noqueados , Proteína 1 Supresora de la Señalización de Citocinas/fisiología , Linfocitos T
4.
BMC Cancer ; 17(1): 157, 2017 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-28235401

RESUMEN

BACKGROUND: Suppressor of cytokine signaling 1 (SOCS1) is considered a tumor suppressor due to frequent epigenetic and micro-RNA-mediated repression of its gene expression in diverse cancers. In prostate cancer (PCa), elevated expression of miR-30d that targets SOCS1 mRNA is associated with increased risk of disease recurrence. SOCS1 can mediate its tumor suppressor functions by diverse mechanisms such as inhibiting the JAK-STAT signaling pathway, promoting the tumor suppressor functions of p53, attenuating MET receptor tyrosine kinase signaling and blocking the oncogenic potential of the cell cycle inhibitor p21CIP1 (p21). Here, we studied the expression of SOCS1 and the downstream targets of its putative tumor suppressor functions (p53, MET and p21) in human PCa specimens to evaluate their significance as markers of disease prognosis. METHODS: Tissue microarrays were constructed of 78 archived prostatectomy specimens that were grouped according to the recommendations of the International Society of Urological Pathology (ISUP) based on the Gleason patterns. SOCS1, p53, MET and p21 protein expression were evaluated by immunohistochemical staining alongside the common prostate cancer-related markers Ki67, prostein and androgen receptor. Statistical correlations between the staining intensities of these markers and ISUP grade groups, local invasion or lymph node metastasis were evaluated. RESULTS: SOCS1 showed diffuse staining in the prostatic epithelium. SOCS1 staining intensity correlated inversely with the ISUP grade groups (ρ = -0.4687, p <0.0001) and Ki67 (ρ = -0.2444, p = 0.031), and positively with prostein (ρ = 0.3511, p = 0.0016). Changes in SOCS1 levels did not significantly associate with those of p53, MET or p21. However, p21 positively correlated with androgen receptor expression (ρ = -0.1388, p = 0.0003). A subset of patients with regional lymph node metastasis, although small in number, showed reduced SOCS1 expression and increased expression of MET and p21. CONCLUSIONS: Our findings suggest that evaluating SOCS1 and p21 protein expression in prostatectomy specimens may have a prognostic value in identifying the aggressive disease. Hence, prospective studies with larger numbers of metastatic PCa specimens incorporating clinical correlates such as disease-free and overall survival are warranted.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/genética , Genes Supresores de Tumor , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo
5.
Cytokine ; 85: 83-91, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27300756

RESUMEN

IL-21 promotes autoimmune type-1 diabetes (T1D) in NOD mice by facilitating CD4(+) T cell help to CD8(+) T cells. IL-21 also enables autoreactive CD8(+) T cells to respond to weak TCR ligands and induce T1D. Here, we assessed whether IL-21 is essential for T1D induction in a mouse model where the disease can occur independently of CD4 help. In this model, which expresses lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) antigen under the rat insulin promoter (RIP-GP), LCMV infection activates CD8(+) T cells reactive to the GP-derived GP33 peptide that attack pancreatic islets and cause T1D. We show that IL-21 deficiency in RIP-GP mice did not impair T1D induction by LCMV expressing the wildtype GP33 peptide. Surprisingly, LCMV-L6F, expressing a weak peptide mimic of GP33, induced T1D more efficiently in Il21(-/-)RIP-GP mice than in controls. However, LCMV-C4Y expressing a very weak peptide mimic of GP33 did not induce T1D in Il21(-/-) mice, but T cells from the infected mice caused disease in lymphopenic RIP-GP mice upon adoptive transfer. Using Nur77(GFP) reporter mice, we show that CD8(+) T cells from Il21(-/-) mice expressing the GP33-specific transgenic P14 TCR showed increased reactivity towards low affinity TCR ligands. Collectively, our findings show that IL-21 is not always required for T1D induction by autoreactive CD8(+) T cells, and suggest that IL-21 may play an important role in regulating CD8(+) T cell reactivity towards low affinity TCR ligands.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucinas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/virología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Insulina/inmunología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/virología , Ligandos , Activación de Linfocitos/inmunología , Recuento de Linfocitos/métodos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos/inmunología , Regiones Promotoras Genéticas/inmunología , Ratas
6.
Cytokine ; 82: 125-39, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26822708

RESUMEN

Hepatocyte growth factor (HGF) signaling via the MET receptor is essential for embryonic development and tissue repair. On the other hand, deregulated MET signaling promotes tumor progression in diverse types of cancers. Even though oncogenic MET signaling remains the major research focus, the HGF-MET axis has also been implicated in diverse aspects of immune cell development and functions. In the presence of other hematopoietic growth factors, HGF promotes the development of erythroid, myeloid and lymphoid lineage cells and thrombocytes. In monocytes and macrophages responding to inflammatory stimuli, induction of autocrine HGF-MET signaling can contribute to tissue repair via stimulating anti-inflammatory cytokine production. HGF-MET signaling can also modulate adaptive immune response by facilitating the migration of Langerhans cells and dendritic cells to draining lymph nodes. However, MET signaling has also been shown to induce tolerogenic dendritic cells in mouse models of graft-versus-host disease and experimental autoimmune encephalomyelitis. HGF-MET axis is also implicated in promoting thymopoiesis and the survival and migration of B lymphocytes. Recent studies have shown that MET signaling induces cardiotropism in activated T lymphocytes. Further understanding of the HGF-MET axis in the immune system would allow its therapeutic manipulation to improve immune cell reconstitution, restore immune homeostasis and to treat immuno-inflammatory diseases.


Asunto(s)
Linfocitos B/inmunología , Factor de Crecimiento de Hepatocito/inmunología , Proteínas Proto-Oncogénicas c-met/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Movimiento Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones
7.
Cytokine ; 82: 58-69, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26748724

RESUMEN

Suppressor of cytokine signaling 1 (SOCS1) is an indispensable regulator of IFNγ signaling and has been implicated in the regulation of liver fibrosis. However, it is not known whether SOCS1 mediates its anti-fibrotic functions in the liver directly, or via modulating IFNγ, which has been implicated in attenuating hepatic fibrosis. Additionally, it is possible that SOCS1 controls liver fibrosis by regulating hepatic stellate cells (HSC), a key player in fibrogenic response. While the activation pathways of HSCs have been well characterized, the regulatory mechanisms are not yet clear. The goals of this study were to dissociate IFNγ-dependent and SOCS1-mediated regulation of hepatic fibrogenic response, and to elucidate the regulatory functions of SOCS1 in HSC activation. Liver fibrosis was induced in Socs1(-/-)Ifng(-/-) mice with dimethylnitrosamine or carbon tetrachloride. Ifng(-/-) and C57BL/6 mice served as controls. Following fibrogenic treatments, Socs1(-/-)Ifng(-/-) mice showed elevated serum ALT levels and increased liver fibrosis compared to Ifng(-/-) mice. The latter group showed higher ALT levels and fibrosis than C57BL/6 controls. The livers of SOCS1-deficient mice showed bridging fibrosis, which was associated with increased accumulation of myofibroblasts and abundant collagen deposition. SOCS1-deficient livers showed increased expression of genes coding for smooth muscle actin, collagen, and enzymes involved in remodeling the extracellular matrix, namely matrix metalloproteinases and tissue inhibitor of metalloproteinases. Primary HSCs from SOCS1-deficient mice showed increased proliferation in response to growth factors such as HGF, EGF and PDGF, and the fibrotic livers of SOCS1-deficient mice showed increased expression of the Pdgfb gene. Taken together, these data indicate that SOCS1 controls liver fibrosis independently of IFNγ and that part of this regulation may occur via regulating HSC proliferation and limiting growth factor availability.


Asunto(s)
Factor de Crecimiento Epidérmico/inmunología , Factor de Crecimiento de Hepatocito/inmunología , Interferón gamma/inmunología , Cirrosis Hepática/inmunología , Factor de Crecimiento Derivado de Plaquetas/inmunología , Transducción de Señal/inmunología , Proteína 1 Supresora de la Señalización de Citocinas/inmunología , Animales , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento de Hepatocito/genética , Interferón gamma/genética , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética
8.
J Neuroinflammation ; 12: 198, 2015 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-26521018

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is an organ-specific autoimmune disease resulting in demyelinating plaques throughout the central nervous system. In MS, the exact role of microglia remains unknown. On one hand, they can present antigens, skew T cell responses, and upregulate the expression of pro-inflammatory molecules. On the other hand, microglia may express anti-inflammatory molecules and inhibit inflammation. Microglia express a wide variety of immune receptors such as nod-like receptors (NLRs). NLRs are intracellular receptors capable of regulating both innate and adaptive immune responses. Among NLRs, Nlrp12 is largely expressed in cells of myeloid origins. It plays a role in immune inflammatory responses by negatively regulating the nuclear factor-kappa B (NF-κB) pathway. Thus, we hypothesize that Nlrp12 suppresses inflammation and ameliorates the course of MS. METHODS: We used experimental autoimmune encephalomyelitis (EAE), a well-characterized mouse model of MS. EAE was induced in wild-type (WT) and Nlrp12 (-/-) mice with myelin oligodendrocyte glycoprotein (MOG):complete Freud's adjuvant (CFA). The spinal cords of healthy and immunized mice were extracted for immunofluorescence and pro-inflammatory gene analysis. Primary murine cortical microglia cell cultures of WT and Nlrp12 (-/-) were prepared with cortices of 1-day-old pups. The cells were stimulated with lipopolysaccharide (LPS) and analyzed for the expression of pro-inflammatory genes as well as pro-inflammatory molecule secretions. RESULTS: Over the course of 9 weeks, the Nlrp12 (-/-) mice demonstrated increased severity in the disease state, where they developed the disease earlier and reached significantly higher clinical scores compared to the WT mice. The spinal cords of immunized WT mice relative to healthy WT mice revealed a significant increase in Nlrp12 messenger ribonucleic acid (mRNA) expression at 1, 3, and 5 weeks post injection. A significant increase in the expression of pro-inflammatory genes Ccr5, Cox2, and IL-1ß was found in the spinal cords of the Nlrp12 (-/-) mice relative to the WT mice (P < 0.05). A significant increase in the level of gliosis was observed in the spinal cords of the Nlrp12 (-/-) mice compared to the WT mice after 9 weeks of disease (P < 0.05). Primary Nlrp12 (-/-) microglia cells demonstrated a significant increase in inducible nitric oxide synthase (iNOS) expression (P < 0.05) and secreted significantly (P < 0.05) more tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), and nitric oxide (NO). CONCLUSION: Nlrp12 plays a protective role by suppressing inflammation during the development of EAE. The absence of Nlrp12 results in an increased inflammatory response.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Animales , Citocinas/biosíntesis , Citocinas/metabolismo , Femenino , Gliosis/genética , Gliosis/patología , Inflamación/genética , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Esclerosis Múltiple/patología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Glicoproteína Oligodendrócito-Mielina/metabolismo , Médula Espinal/inmunología , Médula Espinal/patología , Linfocitos T
9.
Cell Physiol Biochem ; 32(3): 687-98, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080823

RESUMEN

BACKGROUND/AIMS: T-lymphocyte activation and function critically depends on Ca(2+) signaling, which is regulated by store operated Ca(2+) entry (SOCE). Human and mouse T lymphocytes express AMP activated kinase AMPKα1, which is rapidly activated following elevation of cytosolic Ca(2+) concentration ([Ca(2+)]i) by treatment of the cells with Ca(2+) ionophore or following inhibition of endosomal Ca(2+) ATPase with thapsigargin. AMPK is further activated by triggering of the T cell antigen receptor (TCR). The present study explored whether AMPK influences Ca(2+) entry and Ca(2+)-sensitive regulation of T-lymphocyte function. METHODS: T-lymphocytes were isolated and cultured from AMPKα1-deficient (ampk(-/-)) mice and from their wildtype (ampk(+/+)) littermates. The phenotype of the cells was analysed by flow cytometry, [Ca(2+)]i estimated from Fura-2 fluorescence, SOCE from increase of [Ca(2+)]i following thapsigargin treatment (1 µM), and cell function analysed by measuring cytokine secretion and western blotting. RESULTS: Expression of surface markers in CD4(+) and CD8(+) T-cells were similar in ampk(-/-) and ampk(+/+) T-lymphocyte blasts. Moreover, total STIM1 protein abundance was similar in ampk(-/-) and ampk(+/+) T-lymphocyte blasts. However, Orai1 cell membrane protein abundance was significantly higher in ampk(-/-) than in ampk(+/+) T-lymphocyte blasts. SOCE and increase of [Ca(2+)]i following TCR activation by triggering TCR with anti-CD3 and cross-linking secondary antibody were both significantly more pronounced in ampk(-/-) than in ampk(+/+) T-lymphocyte blasts. The difference of Ca(2+) entry between ampk(-/-) and ampk(+/+) T-lymphocytes was abrogated by Orai1 inhibitor 2-aminoethoxydiphenyl borate (2-APB, 50 µM). Proliferation of unstimulated ampk(-/-) lymphocytes was higher than proliferation of ampk(+/+) T-lymphocytes, a difference reversed by Orai1 silencing. CONCLUSIONS: AMPK downregulates Orai1 and thus SOCE in T-lymphocytes and thus participates in negative feed-back regulation of cytosolic Ca(2+) activity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Canales de Calcio/metabolismo , Calcio/metabolismo , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Anticuerpos/inmunología , Compuestos de Boro/farmacología , Complejo CD3/inmunología , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Canales de Calcio/química , Canales de Calcio/genética , Proliferación Celular , Células Cultivadas , Fura-2/química , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Proteína ORAI1 , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Molécula de Interacción Estromal 1 , Tapsigargina/farmacología
10.
Cancers (Basel) ; 15(3)2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36765626

RESUMEN

We previously reported that NOD.Scid mice lacking interleukin-15 (IL-15), or IL-15 receptor alpha-chain, develop T-acute lymphoblastic leukemia (T-ALL). To understand the mechanisms by which IL-15 signaling controls T-ALL development, we studied the thymocyte developmental events in IL-15-deficient Scid mice from NOD and C57BL/6 genetic backgrounds. Both kinds of mice develop T-ALL characterized by circulating TCR-negative cells expressing CD4, CD8 or both. Analyses of thymocytes in NOD.Scid.Il15-/- mice prior to T-ALL development revealed discernible changes within the CD4-CD8- double-negative (DN) thymocyte developmental stages and increased frequencies of CD4+CD8+ double-positive cells with a high proportion of TCR-negative CD4+ and CD8+ cells. The DN cells also showed elevated expressions of CXCR4 and CD117, molecules implicated in the expansion of DN thymocytes. T-ALL cell lines and primary leukemic cells from IL-15-deficient NOD.Scid and C57BL/6.Scid mice displayed increased NOTCH1 activation that was inhibited by NOTCH1 inhibitors and blockers of the PI3K/AKT pathway. Primary leukemic cells from NOD.Scid.Il15-/- mice survived and expanded when cultured with MS5 thymic stromal cells expressing Delta-like ligand 4 and supplemented with IL-7 and FLT3 ligand. These findings suggest that IL-15 signaling in the thymus controls T-ALL development from aberrant thymocytes with an impaired DNA repair capacity and increased NOTCH1 activation.

11.
J Cell Mol Med ; 16(5): 1085-93, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21781276

RESUMEN

Loss-of-function mutations in human adenomatous polyposis coli (APC) lead to multiple colonic adenomatous polyps eventually resulting in colonic carcinoma. Similarly, heterozygous mice carrying defective APC (apc(Min/+)) suffer from intestinal tumours. The animals further suffer from anaemia, which in theory could result from accelerated eryptosis, a suicidal erythrocyte death triggered by enhanced cytosolic Ca(2+) activity and characterized by cell membrane scrambling and cell shrinkage. To explore, whether APC-deficiency enhances eryptosis, we estimated cell membrane scrambling from annexin V binding, cell size from forward scatter and cytosolic ATP utilizing luciferin-luciferase in isolated erythrocytes from apc(Min/+) mice and wild-type mice (apc(+/+)). Clearance of circulating erythrocytes was estimated by carboxyfluorescein-diacetate-succinimidyl-ester labelling. As a result, apc(Min/+) mice were anaemic despite reticulocytosis. Cytosolic ATP was significantly lower and annexin V binding significantly higher in apc(Min/+) erythrocytes than in apc(+/+) erythrocytes. Glucose depletion enhanced annexin V binding, an effect significantly more pronounced in apc(Min/+) erythrocytes than in apc(+/+) erythrocytes. Extracellular Ca(2+) removal or inhibition of Ca(2+) entry with amiloride (1 mM) blunted the increase but did not abrogate the genotype differences of annexin V binding following glucose depletion. Stimulation of Ca(2+) -entry by treatment with Ca(2+) -ionophore ionomycin (10 µM) increased annexin V binding, an effect again significantly more pronounced in apc(Min/+) erythrocytes than in apc(+/+) erythrocytes. Following retrieval and injection into the circulation of the same mice, apc(Min/+) erythrocytes were more rapidly cleared from circulating blood than apc(+/+) erythrocytes. Most labelled erythrocytes were trapped in the spleen, which was significantly enlarged in apc(Min/+) mice. The observations point to accelerated eryptosis and subsequent clearance of apc(Min/+) erythrocytes, which contributes to or even accounts for the enhanced erythrocyte turnover, anaemia and splenomegaly in those mice.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Apoptosis/genética , Eritrocitos/fisiología , Genes APC , Mutación , Amilorida/farmacología , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Ionóforos de Calcio/farmacología , Membrana Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/metabolismo , Eritrocitos/efectos de los fármacos , Femenino , Ionomicina/farmacología , Masculino , Ratones , Fosfatidilserinas/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Bazo/fisiología
12.
Cell Physiol Biochem ; 29(3-4): 533-42, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22508060

RESUMEN

Azathioprine is a powerful immunosuppressive drug, which is partially effective by interfering with the maturation and function of dendritic cells (DCs), antigen-presenting cells linking innate and adaptive immunity. DCs are stimulated by bacterial lipopolysaccharides (LPS), which trigger the formation of reactive oxygen species (ROS), paralleled by activation of the Na(+)/H(+) exchanger. The carrier is involved in the regulation of cytosolic pH, cell volume and migration. The present study explored whether azathioprine influences Na(+)/H(+) exchanger activity in DCs. DCs were isolated from murine bone marrow, cytosolic pH (pH(i)) was estimated utilizing 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF-AM) fluorescence, Na(+)/H(+) exchanger activity from the Na(+)-dependent realkalinization following an ammonium pulse, cell volume from forward scatter in FACS analysis, ROS production from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence, TNFα release utilizing ELISA, and migration utilizing transwell migration assays. Exposure of DCs to lipopolysaccharide (LPS, 1 µg/ml) led to a transient increase of Na(+)/H(+) exchanger activity, an effect paralleled by ROS formation, increased cell volume, TNFα production and stimulated migration. Azathioprine (10 µM) did not significantly alter the Na(+)/H(+) exchanger activity, cell volume and ROS formation prior to LPS exposure but significantly blunted the LPS-induced stimulation of Na(+)/H(+) exchanger activity, ROS formation, cell swelling, TNFα production and cell migration. In conclusion, azathioprine interferes with the activation of dendritic cell Na(+)/H(+) exchanger by bacterial lipopolysaccharides, an effect likely participating in the anti-inflammatory action of the drug.


Asunto(s)
Azatioprina/farmacología , Células Dendríticas/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/efectos de los fármacos , Animales , Movimiento Celular , Tamaño de la Célula , Células Cultivadas , Citosol/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/fisiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Fluoresceínas/metabolismo , Fluorescencia , Concentración de Iones de Hidrógeno , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Intercambiadores de Sodio-Hidrógeno/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Cell Physiol Biochem ; 29(3-4): 543-50, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22508061

RESUMEN

Rapamycin, a widely used immunosuppressive drug, has been shown to interfere with the function of dendritic cells (DCs), antigen-presenting cells contributing to the initiation of primary immune responses and the establishment of immunological memory. DC function is governed by the Na(+)/H(+) exchanger (NHE), which is activated by bacterial lipopolysaccharides (LPS) and is required for LPS-induced cell swelling, reactive oxygen species (ROS) production and TNF-α release. The present study explored, whether rapamycin influences NHE activity and/or ROS formation in DCs. Mouse DCs were treated with LPS in the absence and presence of rapamycin (100 nM). ROS production was determined from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence, cytosolic pH (pH(i)) from 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) fluorescence, NHE activity from the Na(+)-dependent realkalinization following an ammonium pulse, cell volume from forward scatter in FACS analysis, and TNF-α production utilizing ELISA. In the absence of LPS, rapamycin did not significantly modify cytosolic pH, NHE activity or cell volume but significantly decreased ROS formation. LPS stimulated NHE activity, enhanced forward scatter, increased ROS formation, and triggered TNF-α release, effects all blunted in the presence of rapamycin. NADPH oxidase inhibitor Vas-2870 (10 µM) mimicked the effect of rapamycin on LPS induced stimulation of NHE activity and TNF-α release. The effect of rapamycin on TNF-α release was also mimicked by the antioxidant ROS scavenger Tempol (30 µM) and partially reversed by additional application of tert-butylhydroperoxide (10 µM). In conclusion, in DCs rapamycin disrupts LPS induced ROS formation with subsequent inhibition of NHE activity, cell swelling and TNF-α release.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Sirolimus/farmacología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Benzoxazoles/farmacología , Tamaño de la Célula , Células Cultivadas , Óxidos N-Cíclicos/farmacología , Citosol/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/fisiología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/química , Citometría de Flujo , Fluoresceínas/metabolismo , Fluorescencia , Concentración de Iones de Hidrógeno , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Intercambiadores de Sodio-Hidrógeno/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/fisiología , Marcadores de Spin , Triazoles/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , terc-Butilhidroperóxido/farmacología
14.
Cell Physiol Biochem ; 29(1-2): 21-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22415071

RESUMEN

The anti-inflammatory Nigella sativa component thymoquinone compromises the function of dendritic cells (DCs), key players in the regulation of innate and adaptive immunity. DC function is regulated by the Na(+)/H(+) exchanger (NHE), which is stimulated by lipopolysaccharides (LPS) and required for LPS-induced cell swelling, reactive oxygen species (ROS) production, TNF-α release and migration. Here we explored, whether thymoquinone influences NHE activity in DCs. To this end, bone marrow derived mouse DCs were treated with LPS in the absence and presence of thymoquinone (10 µM). Cytosolic pH (pH(i)) was determined from 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) fluorescence, NHE activity from the Na(+)-dependent realkalinization following an ammonium pulse, cell volume from forward scatter in FACS analysis, ROS production from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence, TNF-α production utilizing ELISA and DC migration with transwell migration assays. As a result, exposure of DCs to LPS (1 µg/ml) led within 4 hours to transient increase of NHE activity. Thymoquinone did not significantly modify cytosolic pH or cellular NHE activity in the absence of LPS, but abrogated the effect of LPS on NHE activity. Accordingly, in the presence of thymoquinone LPS-treatment resulted in cytosolic acidification. LPS further increased forward scatter and ROS formation, effects similarly abrogated by thymoquinone. Again, in the absence of LPS, thymoquinone did not significantly modify ROS formation and cell volume. LPS further triggered TNF-α release and migration, effects again blunted in the presence of thymoquinone. NHE1 inhibitor cariporide (10 µM) blunted LPS induced TNF-α release and migration. The effects of thymoquinone on NHE activity and migration were reversed upon treatment of the cells with t-butyl hydroperoxide (TBOOH, 5 µM). In conclusion, thymoquinone blunts LPS induced NHE activity, cell swelling, oxidative burst, cytokine release and migration of bone marrow derived murine dendritic cells. NHE inhibition may thus contribute to the antiinflammatory action of thymoquinone.


Asunto(s)
Antiinflamatorios/farmacología , Benzoquinonas/farmacología , Células Dendríticas/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Fluoresceínas/química , Guanidinas/farmacología , Concentración de Iones de Hidrógeno/efectos de los fármacos , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Sulfonas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , terc-Butilhidroperóxido/farmacología
15.
Biochem Biophys Res Commun ; 425(1): 6-12, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22814108

RESUMEN

Survival and function of T-lymphocytes critically depends on phosphoinositide (PI) 3 kinase. PI3 kinase signaling includes the PKB/Akt and SGK dependent phosphorylation and thus inhibition of glycogen synthase kinase GSK3α,ß. Lithium, a known unspecific GSK3 inhibitor protects against experimental autoimmune encephalomyelitis. The present study explored, whether Akt/SGK-dependent regulation of GSK3 activity is a determinant of T cell survival and function. Experiments were performed in mutant mice in which Akt/SGK-dependent GSK3α,ß inhibition was disrupted by replacement of the serine residue in the respective SGK/Akt-phosphorylation consensus sequence by alanine (gsk3(KI)). T cells from gsk3(KI) mice were compared to T cells from corresponding wild type mice (gsk3(WT)). As a result, in gsk3(KI) CD4(+) cells surface CD62L (L-selectin) was significantly less abundant than in gsk3(WT) CD4(+) cells. Upon activation in vitro T cells from gsk3(KI) mice reacted with enhanced perforin production and reduced activation induced cell death. Cytokine production was rather reduced in gsk3(KI) T cells, suggesting that GSK3 induces effector function in CD8(+) T cells. In conclusion, PKB/Akt and SGK sensitive phosphorylation of GSK3α,ß is a potent regulator of perforin expression and activation induced cell death in T lymphocytes.


Asunto(s)
Apoptosis/inmunología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Selectina L/biosíntesis , Activación de Linfocitos , Perforina/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Autoinmunidad , Técnicas de Sustitución del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Mutantes , Fosforilación , Linfocitos T Citotóxicos/enzimología
16.
Cell Physiol Biochem ; 27(5): 547-56, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691072

RESUMEN

Janus kinase 3, a tyrosine kinase expressed in haematopoetic tissues, plays a decisive role in T-lymphocyte survival. JAK3 deficiency leads to (Severe) Combined Immunodeficiency (SCID) resulting from enhanced lymphocyte apoptosis. JAK3 is activated by phosphorylation. Nothing is known about expression of JAK3 in erythrocytes, which may undergo apoptosis-like cell death (eryptosis) characterized by cell membrane scrambling with phosphatidylserine exposure and cell shrinkage. Triggers of eryptosis include energy depletion. The present study utilized immunohistochemistry and confocal microscopy to test for JAK3 expression and phosphorylation, and FACS analysis to determine phosphatidylserine exposure (annexin binding) and cell volume (forward scatter). As a result, JAK3 was expressed in erythrocytes and phosphorylated following 24h and 48h glucose depletion. Forward scatter was slightly but significantly smaller in erythrocytes from JAK3-deficient mice (jak3(-/-)) than in erythrocytes from wild type mice (jak3(+/+)). Annexin V binding was similarly low in both genotypes. The JAK3 inhibitors WHI-P131/JANEX-1 (4-(4'-Hydroxyphenyl)amino-6,7-dimethoxyquinazoline, 156µM) and WHI-P154 (4-[(3'-Bromo-4'-hydroxyphenyl)amino]-6,7-dimethoxyquinazoline, 11.2µM) did not significantly modify annexin V binding or forward scatter. Glucose depletion increased annexin V binding, an effect significantly blunted in jak3(-/-) erythrocytes and in the presence of the JAK3 inhibitors. The observations disclose a completely novel role of Janus kinase 3, i.e. the triggering of cell membrane scrambling in energy depleted erythrocytes.


Asunto(s)
Eritrocitos/enzimología , Glucosa/farmacología , Janus Quinasa 3 , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/análisis , Adenosina Trifosfato/metabolismo , Animales , Anexina A5/análisis , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Tamaño de la Célula , Recuento de Eritrocitos , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Femenino , Citometría de Flujo , Eliminación de Gen , Glucosa/deficiencia , Humanos , Inmunohistoquímica , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/biosíntesis , Janus Quinasa 3/deficiencia , Janus Quinasa 3/genética , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Fosfatidilserinas/análisis , Fosfatidilserinas/metabolismo , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología
17.
Cell Physiol Biochem ; 28(2): 347-54, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21865743

RESUMEN

Benzethonium, an antimicrobial surfactant widely used as preservative of pharmaceuticals, topical wound care product and oral disinfectant, triggers apoptosis of several cell types. The apoptosis is preceded and possibly triggered by mitochondrial depolarization. Even though lacking mitochondria, erythrocytes may similarly undergo suicidal cell death or eryptosis. Hallmarks of eryptosis include cell shrinkage and cell membrane scrambling with phosphatidylserine exposure at the cell surface. Eryptosis may be triggered by energy depletion, which leads to increase of cytosolic Ca(2+)-activity with subsequent Ca(2+)-sensitive cell shrinkage and cell membrane scrambling. Ca(2+)-sensitivity is enhanced by ceramide. The present study explored the effect of benzethonium on eryptosis. Cell membrane scrambling was estimated from binding of fluorescent annexin V to phosphatidylserine, cell volume from forward scatter in FACS analysis, cytosolic Ca(2+)-concentration from Fluo3-fluorescence, hemolysis from hemoglobin release, lactate formation by colorimetry and ceramide utilizing fluorescent antibodies. A 48 hours exposure to benzethonium (=5µM) significantly increased cytosolic Ca(2+)-concentration, decreased forward scatter and triggered annexin V-binding affecting some 30% of the erythrocytes at 5 µM benzethonium. Only 5% of treated erythrocytes were hemolytic. The effects of benzethonium on annexin V binding were blunted in the nominal absence of Ca(2+) and in the presence of amiloride (1 mM) but not in the presence of the pancaspase inhibitor zVAD (10 µM). Benzethonium further significantly enhanced the effect of glucose depletion on cytosolic Ca(2+)-concentration and annexin V-binding, but significantly blunted the effect of glucose depletion on forward scatter. Benzethonium (5 µM) significantly enhanced lactic acid formation but not ceramide abundance. The present observations disclose a novel effect of benzethonium, i.e. triggering of suicidal death of erythrocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Bencetonio/farmacología , Eritrocitos/efectos de los fármacos , Amilorida/farmacología , Compuestos de Anilina/química , Anexina A5/metabolismo , Calcio/metabolismo , Ceramidas/metabolismo , Eritrocitos/metabolismo , Eritrocitos/fisiología , Citometría de Flujo , Colorantes Fluorescentes/química , Hemólisis , Humanos , Oligopéptidos/farmacología , Fosfatidilserinas/farmacología , Unión Proteica , Xantenos/química
18.
Cell Physiol Biochem ; 27(1): 45-54, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21325821

RESUMEN

The preclinical compounds Bay 11-7082 and parthenolide trigger apoptosis, an effect contributing to their antiinflammatory action. The substances interfere with the activation and nuclear translocation of nuclear factor NFκB, by inhibiting NFκB directly (parthenolide) or by interfering with the inactivation of the NFκB inhibitory protein IκB-α (Bay 11-7082). Beyond that, the substances may be effective in part by nongenomic effects. Similar to apoptosis of nucleated cells, erythrocytes may undergo apoptosis-like cell death (eryptosis) characterized by cell membrane scrambling with phosphatidylserine exposure, and cell shrinkage. Thus, erythrocytes allow the study of nongenomic mechanisms contributing to suicidal cell death, e.g. Ca(2+) leakage or glutathione depletion. The present study utilized Western blotting to search for NFκB and IκB-α expression in erythrocytes, FACS analysis to determine cytosolic Ca(2+) (Fluo3 fluorescence), phosphatidylserine exposure (annexin V binding), and cell volume (forward scatter), as well as an enzymatic method to determine glutathione levels. As a result, both NFκB and IκB-α are expressed in erythrocytes. Targeting the NFκB pathway by Bay 11-7082 (IC(50) ≈ 10 µM) and parthenolide (IC(50) ≈ 30 µM) triggered suicidal erythrocyte death as shown by annexin V binding and decrease of forward scatter. Bay 11-7082 treatment further increased intracellular Ca(2+) and led to depletion of reduced glutathione. The effects of Bay 11-7082 and parthenolide on annexin V binding could be fully reversed by the antioxidant N-acetylcysteine. In conclusion, the pharmacological inhibitors of NFκB, Bay 11-7082 and parthenolide, interfere with the survival of erythrocytes involving mechanisms other than disruption of NFκB-dependent gene expression.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis , Eritrocitos/metabolismo , FN-kappa B/metabolismo , Nitrilos/farmacología , Sesquiterpenos/farmacología , Sulfonas/farmacología , Compuestos de Anilina/química , Anexina A5/metabolismo , Calcio/metabolismo , Tamaño de la Célula , Eritrocitos/efectos de los fármacos , Glutatión/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , FN-kappa B/antagonistas & inhibidores , Fosfatidilserinas/metabolismo , Unión Proteica , Transducción de Señal , Xantenos/química
19.
FASEB J ; 24(9): 3351-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20427706

RESUMEN

Endothelins (ETs), potent endothelium-derived mediators, stimulate formation of nitric oxide, which, in turn, protects against suicidal erythrocyte death or eryptosis, characterized by phosphatidylserine exposure at the erythrocyte surface and triggered by increase in cytosolic Ca(2+) ([Ca(2+)](i)). The present study explored whether the ET1-receptor ETB influences suicidal erythrocyte death. To this end, [Ca(2+)](i) (Fluo3-fluorescence) and phosphatidylserine exposure (annexin V-binding) were determined utilizing FACS analysis. Energy depletion increased [Ca(2+)](i) and phosphatidylserine-exposure, effects significantly blunted by ET1 (IC(50) approximately 100 nM) and the ETB receptor-agonist sarafotoxin 6c (IC(50) approximately 10 nM) but not by ET2 and ET3. ET1 and sarafotoxin significantly delayed the kinetics of suicidal erythrocyte death following energy depletion. ETB stimulation did not blunt the effect of Ca(2+)-ionophore ionomycin (1 microM) on phosphatidylserine exposure. The in vivo significance was tested using rescued ETB-knockout (etb(-/-)) and wild-type (etb(+/+)) mice. The number of phosphatidylserine-exposing erythrocytes, of reticulocytes and spleen size were significantly larger in etb(-/-) mice than in etb(+/+)-mice. The etb(-/-) erythrocytes were more susceptible to the eryptotic effect of oxidative stress and more rapidly cleared from circulating blood than etb(+/+) erythrocytes. Finally, the spleens from etb(-/-) mice were enlarged and contained markedly more phosphatidylserine-exposing erythrocytes than spleens from etb(+/+) mice. The observations disclose a novel function of ET1, i.e., protection from suicidal erythrocyte death.


Asunto(s)
Eritrocitos/citología , Eritrocitos/metabolismo , Receptor de Endotelina B/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Endotelina-1/farmacología , Eritrocitos/efectos de los fármacos , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ionomicina/farmacología , Ionóforos/farmacología , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Receptor de Endotelina B/agonistas , Receptor de Endotelina B/genética , Venenos de Víboras/farmacología
20.
Malar J ; 10: 139, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21599958

RESUMEN

BACKGROUND: Gum Arabic (GA), a nonabsorbable nutrient from the exudate of Acacia senegal, exerts a powerful immunomodulatory effect on dendritic cells, antigen-presenting cells involved in the initiation of both innate and adaptive immunity. On the other hand GA degradation delivers short chain fatty acids, which in turn have been shown to foster the expression of foetal haemoglobin in erythrocytes. Increased levels of erythrocyte foetal haemoglobin are known to impede the intraerythrocytic growth of Plasmodium and thus confer some protection against malaria. The present study tested whether gum arabic may influence the clinical course of malaria. METHODS: Human erythrocytes were in vitro infected with Plasmodium falciparum in the absence and presence of butyrate and mice were in vivo infected with Plasmodium berghei ANKA by injecting parasitized murine erythrocytes (1 × 106) intraperitoneally. Half of the mice received gum arabic (10% in drinking water starting 10 days before the day of infection). RESULTS: According to the in vitro experiments butyrate significantly blunted parasitaemia only at concentrations much higher (3 mM) than those encountered in vivo following GA ingestion (<1 µM). According to the in vivo experiments the administration of gum arabic slightly but significantly decreased the parasitaemia and significantly extended the life span of infected mice. DISCUSSION: GA moderately influences the parasitaemia and survival of Plasmodium-infected mice. The underlying mechanism remained, however, elusive. CONCLUSIONS: Gum arabic favourably influences the course of murine malaria.


Asunto(s)
Antimaláricos/administración & dosificación , Goma Arábiga/administración & dosificación , Malaria/tratamiento farmacológico , Administración Oral , Animales , Antimaláricos/farmacología , Modelos Animales de Enfermedad , Femenino , Goma Arábiga/farmacología , Experimentación Humana , Humanos , Masculino , Ratones , Parasitemia/tratamiento farmacológico , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/patogenicidad , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Enfermedades de los Roedores/tratamiento farmacológico , Enfermedades de los Roedores/parasitología , Análisis de Supervivencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA