Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Front Immunol ; 14: 1111797, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817433

RESUMEN

Background: COVID-19 severity has been linked to an increased production of inflammatory mediators called "cytokine storm". Available data is mainly restricted to the first international outbreak and reports highly variable results. This study compares demographic and clinical features of patients with COVID-19 from Córdoba, Argentina, during the first two waves of the pandemic and analyzes association between comorbidities and disease outcome with the "cytokine storm", offering added value to the field. Methods: We investigated serum concentration of thirteen soluble mediators, including cytokines and chemokines, in hospitalized patients with moderate and severe COVID-19, without previous rheumatic and autoimmune diseases, from the central region of Argentina during the first and second infection waves. Samples from healthy controls were also assayed. Clinical and biochemical parameters were collected. Results: Comparison between the two first COVID-19 waves in Argentina highlighted that patients recruited during the second wave were younger and showed less concurrent comorbidities than those from the first outbreak. We also recognized particularities in the signatures of systemic cytokines and chemokines in patients from both infection waves. We determined that concurrent pre-existing comorbidities did not have contribution to serum concentration of systemic cytokines and chemokines in COVID-19 patients. We also identified immunological and biochemical parameters associated to inflammation which can be used as prognostic markers. Thus, IL-6 concentration, C reactive protein level and platelet count allowed to discriminate between death and discharge in patients hospitalized with severe COVID-19 only during the first but not the second wave. Conclusions: Our data provide information that deepens our understanding of COVID-19 pathogenesis linking demographic features of a COVID-19 cohort with cytokines and chemokines systemic concentration, presence of comorbidities and different disease outcomes. Altogether, our findings provide information not only at local level by delineating inflammatory/anti-inflammatory response of patients but also at international level addressing the impact of comorbidities and the infection wave in the variability of cytokine and chemokine production upon SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Humanos , Citocinas/metabolismo , SARS-CoV-2/metabolismo , Argentina , Quimiocinas , Síndrome de Liberación de Citoquinas , Pandemias
2.
Brain Behav Immun ; 101: 359-376, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35065197

RESUMEN

Stressful experience-induced cocaine-related behaviors are associated with a significant impairment of glutamatergic mechanisms in the Nucleus Accumbens core (NAcore). The hallmarks of disrupted glutamate homeostasis following restraint stress are the enduring imbalance of glutamate efflux after a cocaine stimulus and increased basal concentrations of extracellular glutamate attributed to GLT-1 downregulation in the NAcore. Glutamate transmission is tightly linked to microglia functioning. However, the role of microglia in the biological basis of stress-induced addictive behaviors is still unknown. By using minocycline, a potent inhibitor of microglia activation with anti-inflammatory properties, we determined whether microglia could aid chronic restraint stress (CRS)-induced glutamate homeostasis disruption in the NAcore, underpinning stress-induced cocaine self-administration. In this study, adult male rats were restrained for 2 h/day for seven days (day 1-7). From day 16 until completing the experimental protocol, animals received a vehicle or minocycline treatment (30 mg/Kg/12h i.p.). On day 21, animals were assigned to microscopic, biochemical, neurochemical or behavioral studies. We confirm that the CRS-induced facilitation of cocaine self-administration is associated with enduring GLT-1 downregulation, an increase of basal extracellular glutamate and postsynaptic structural plasticity in the NAcore. These alterations were strongly related to the CRS-induced reactive microglia and increased TNF-α mRNA and protein expression, since by administering minocycline, the impaired glutamate homeostasis and the facilitation of cocaine self-administration were prevented. Our findings are the first to demonstrate that minocycline suppresses the CRS-induced facilitation of cocaine self-administration and glutamate homeostasis disruption in the NAcore. A role of microglia is proposed for the development of glutamatergic mechanisms underpinning stress-induced vulnerability to cocaine addiction.


Asunto(s)
Cocaína , Animales , Cocaína/metabolismo , Ácido Glutámico/metabolismo , Masculino , Microglía/metabolismo , Minociclina/metabolismo , Minociclina/farmacología , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley
3.
Int Immunopharmacol ; 105: 108546, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35074570

RESUMEN

The development of neuroinflammation, as well as the progression of several neurodegenerative diseases, has been associated with the activation and mobilization of the peripheral immune system due to systemic inflammation. However, the mechanism by which this occurs remains unclear. Here, we addressed the effect of systemic sterile induced-co-expression of IL-12 and IL-18, in the establishment of a novel cytokine-mediated model of neuroinflammation. Following peripheral hydrodynamic shear of IL-12 plus IL-18 cDNAs in C57BL/6 mice, we induced systemic and persistent level of IL-12, which in turn promoted the elevation of circulating pro-inflammatory cytokines TNF-α and IFN-γ, accompanied with splenomegaly. Moreover, even though we identified an increased gene expression of both TNF-α and IFN-γ in the brain, we observed that only IFN-γ, but not TNF-α signaling through its type I receptor, was required to induce both the trafficking of leukocytes from the periphery toward the brain and upregulate MHC-II in microglia and inflammatory monocytes. Therefore, only TNF-α was shown to be dispensable, revealing an IFN-γ-dependent activation of microglia and recruitment of leukocytes, particularly of highly activated inflammatory monocytes. Taken together, our results argue for a systemic cytokine-mediated establishment and development of neuroinflammation, having identified IFN-γ as a potential target for immunomodulation.


Asunto(s)
Interferón gamma , Microglía , Animales , Encéfalo/metabolismo , Citocinas/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Monocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-32774323

RESUMEN

Chronic lymphocytic leukemia (CLL) is the most common type of adult leukemia in the western hemisphere. It is characterized by a clonal proliferation of a population of CD5+ B lymphocytes that accumulate in the secondary lymphoid tissues, bone marrow, and blood. Some CLL patients remain free of symptoms for decades, whereas others rapidly become symptomatic or develop high-risk disease. Studying autophagy, which may modulate key protein expression and cell survival, may be important to the search for novel prognostic factors and molecules. Here, we applied flow cytometry technology to simultaneously detect autophagy protein LC3B with classical phenotypical markers used for the identification of tumoral CLL B cell clones. We found that two patients with progressing CLL showed increased expression of the autophagy protein LC3B, in addition to positive expression of CD38 and ZAP70 and unmutated status of IGHV. Our data suggest that activation of autophagy flux may correlate with CLL progression even before Ibrutinib treatment.


Asunto(s)
Autofagia , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Adulto , Progresión de la Enfermedad , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico
5.
Front Immunol ; 10: 80, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30761145

RESUMEN

Innate immune activation and chronic neuroinflammation are characteristic features of many neurodegenerative diseases including Parkinson's disease (PD) and may contribute to the pathophysiology of the disease. The discovery of misfolded alpha-synuclein (αSYN) protein aggregates, which amplify in a "prion-like" fashion, has led us to consider that pathogenic αSYN might be hijacking the activation and mobilization mechanism of the peripheral immune system to reach and disseminate within the CNS. Furthermore, our lab and other groups have recently shown that αSYN can adopt distinct fibril conformations or "strains" with varying levels of pathogenic impact. Therefore, the aim of this study was to assess the impact of peripheral inflammation on αSYN spreading in order to better understand the participation of the immune system in the progression of PD. The results presented here show that intraperitoneal LPS injection prior to systemic intravenous recombinant administration of two different αSYN pathogenic strains (fibrils or ribbons) in wild type mice, induces an increase in brain resident microglia and promotes the recruitment of leukocytes toward the brain and the spinal cord. Our findings show for the first time that αSYN can be internalized by LPS-primed inflammatory monocytes, which in turn favors the dissemination from the periphery toward the brain and spinal cord. Further, we found a differential recruitment of CD4+ and CD8+ T cells after LPS priming and subsequent administration of the αSYN ribbons strain. Together, these data argue for a role of the peripheral immune system in αSYN pathology.


Asunto(s)
Encéfalo/inmunología , Vigilancia Inmunológica , Inflamación/inmunología , Monocitos/metabolismo , Médula Espinal/inmunología , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacología , Administración Intravenosa , Animales , Encéfalo/patología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Inyecciones Intraperitoneales , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Enfermedad de Parkinson/inmunología , Agregado de Proteínas , Médula Espinal/patología , alfa-Sinucleína/administración & dosificación
7.
J Cell Sci ; 131(23)2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30404831

RESUMEN

Autophagic dysfunction and protein aggregation have been linked to several neurodegenerative disorders, but the exact mechanisms and causal connections are not clear and most previous work was done in neurons and not in microglial cells. Here, we report that exogenous fibrillary, but not monomeric, alpha-synuclein (AS, also known as SNCA) induces autophagy in microglial cells. We extensively studied the dynamics of this response using both live-cell imaging and correlative light-electron microscopy (CLEM), and found that it correlates with lysosomal damage and is characterised by the recruitment of the selective autophagy-associated proteins TANK-binding kinase 1 (TBK1) and optineurin (OPTN) to ubiquitylated lysosomes. In addition, we observed that LC3 (MAP1LC3B) recruitment to damaged lysosomes was dependent on TBK1 activity. In these fibrillar AS-treated cells, autophagy inhibition impairs mitochondrial function and leads to microglial cell death. Our results suggest that microglial autophagy is induced in response to lysosomal damage caused by persistent accumulation of AS fibrils. Importantly, triggering of the autophagic response appears to be an attempt at lysosomal quality control and not for engulfment of fibrillar AS.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Lisosomas/metabolismo , Microglía/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción TFIIIA/genética , alfa-Sinucleína/metabolismo , Autofagia , Proteínas de Ciclo Celular , Humanos , Proteínas de Transporte de Membrana , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factor de Transcripción TFIIIA/metabolismo
8.
Front Immunol ; 9: 770, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29719536

RESUMEN

Acute brain injury leads to the recruitment and activation of immune cells including resident microglia and infiltrating peripheral myeloid cells (MC), which contribute to the inflammatory response involved in neuronal damage. We previously reported that TLR2 stimulation by peptidoglycan (PGN) from Staphylococcus aureus, in vitro and in vivo, induced microglial cell activation followed by autophagy induction. In this report, we evaluated if phosphatidyl-inositol-3 kinase (PI3K) pharmacological inhibitors LY294200 and 3-methyladenine (3-MA) can modulate the innate immune response to PGN in the central nervous system. We found that injection of PGN into the mouse brain parenchyma (caudate putamen) triggered an inflammatory reaction, which involved activation of microglial cells, recruitment of infiltrating MC to injection site, production of pro-inflammatory mediators, and neuronal injury. In addition, we observed the accumulation of LC3B+ CD45+ cells and colocalization of LC3B and lysosomal-associated membrane protein 1 in brain cells. Besides, we found that pharmacological inhibitors of PI3K, including the classical autophagy inhibitor 3-MA, reduced the recruitment of MC, microglial cell activation, and neurotoxicity induced by brain PGN injection. Collectively, our results suggest that PI3K pathways and autophagic response may participate in the PGN-induced microglial activation and MC recruitment to the brain. Thus, inhibition of these pathways could be therapeutically targeted to control acute brain inflammatory conditions.


Asunto(s)
Encéfalo/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Inflamación/inmunología , Peptidoglicano/toxicidad , Inhibidores de las Quinasa Fosfoinosítidos-3 , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Encéfalo/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Inhibidores Enzimáticos/farmacología , Inflamación/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo
10.
Front Immunol ; 8: 1666, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29255461

RESUMEN

Brain-resident microglia and peripheral migratory leukocytes play essential roles in shaping the immune response in the central nervous system. These cells activate and migrate in response to chemokines produced during active immune responses and may contribute to the progression of neuroinflammation. Herein, we addressed the participation of type I-II interferons in the response displayed by microglia and inflammatory monocytes to comprehend the contribution of these cytokines in the establishment and development of a neuroinflammatory process. Following systemic lipopolysaccharide (LPS) challenge, we found glial reactivity and an active recruitment of CD45hi leukocytes close to CD31+ vascular endothelial cells in circumventricular organs. Isolated CD11b+ CD45hi Ly6Chi Ly6G--primed inflammatory monocytes were able to induce T cell proliferation, unlike CD11b+ CD45lo microglia. Moreover, ex vivo re-stimulation with LPS exhibited an enhancement of T cell proliferative response promoted by inflammatory monocytes. These myeloid cells also proved to be recruited in a type I interferon-dependent fashion as opposed to neutrophils, unveiling a role of these cytokines in their trafficking. Together, our results compares the phenotypic and functional features between tissue-resident vs peripheral recruited cells in an inflamed microenvironment, identifying inflammatory monocytes as key sentinels in a LPS-induced murine model of neuroinflammation.

12.
Sci Rep ; 7: 43153, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28256519

RESUMEN

Autophagy is a fundamental cellular homeostatic mechanism, whereby cells autodigest parts of their cytoplasm for removal or turnover. Neurodegenerative disorders are associated with autophagy dysregulation, and drugs modulating autophagy have been successful in several animal models. Microglial cells are phagocytes in the central nervous system (CNS) that become activated in pathological conditions and determine the fate of other neural cells. Here, we studied the effects of autophagy on the production of pro-inflammatory molecules in microglial cells and their effects on neuronal cells. We observed that both trehalose and rapamycin activate autophagy in BV2 microglial cells and down-regulate the production of pro-inflammatory cytokines and nitric oxide (NO), in response to LPS and alpha-synuclein. Autophagy also modulated the phosphorylation of p38 and ERK1/2 MAPKs in BV2 cells, which was required for NO production. These actions of autophagy modified the impact of microglial activation on neuronal cells, leading to suppression of neurotoxicity. Our results demonstrate a novel role for autophagy in the regulation of microglial cell activation and pro-inflammatory molecule secretion, which may be important for the control of inflammatory responses in the CNS and neurotoxicity.


Asunto(s)
Autofagia , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Lipopolisacáridos/toxicidad , Neuroglía/fisiología , Óxido Nítrico/metabolismo , alfa-Sinucleína/toxicidad , Animales , Línea Celular , Ratones , Transducción de Señal
13.
PLoS One ; 9(2): e90116, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587231

RESUMEN

For more than a decade, the cytokine interleukin-12 (IL-12) has been utilized, either alone or in combination with other drugs, as a treatment for cancer. The numerous anti-tumor properties of IL-12 still generate interest in the clinical use of this cytokine, even though it has demonstrated toxicity when administrated systemically. As an approach to overcome this toxicity, numerous laboratories have attempted to induce IL-12 expression at the site of the tumor. However for tumors that are difficult to remove surgically or for the treatment of disseminated metastases, systemic expression of this cytokine still remains as the most efficient method of administration. Nevertheless, finding alternative approaches for the use of IL-12 in the treatment of cancer and unraveling the basis of IL-12-side effects remain a challenge. In the present work we demonstrate that systemic expression of IL-12 through hydrodynamic injection of IL-12 cDNA is able to induce different types of liver lesions associated with a toxic pathology. However we report here that hepatic toxicity is diminished and survival of mice enhanced in the absence of tumor necrosis factor alpha (TNFα). This observation is in contrast to several murine models and clinical trials that postulate interferon gamma (IFNγ) as the main cytokine responsible for IL-12 toxicity. Moreover, our work demonstrates that when IL-12 cDNA is co-injected with IL-18 cDNA or when mice are pre-treated with a low dose of IL-12 cDNA prior to receiving a high dose of IL-12 cDNA, systemic levels of TNFα are almost completely abrogated, resulting in improved survival and less hepatic damage. Importantly, abrogation of TNFα signaling does not affect the strong anti-tumor activity of IL-12. Thus, neutralizing TNFα with antagonists already approved for human use offers a promising approach to abrogate IL-12 side effects during the use of this cytokine for the treatment of cancer.


Asunto(s)
ADN Complementario/administración & dosificación , Inmunoterapia/métodos , Interleucina-12/inmunología , Interleucina-18/inmunología , Melanoma Experimental/terapia , Neoplasias del Bazo/terapia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , ADN Complementario/inmunología , Expresión Génica , Hidrodinámica , Inyecciones Intravenosas , Interleucina-12/biosíntesis , Interleucina-12/genética , Interleucina-18/biosíntesis , Interleucina-18/genética , Hígado/efectos de los fármacos , Hígado/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias del Bazo/inmunología , Neoplasias del Bazo/patología , Cola (estructura animal) , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
Int Immunopharmacol ; 18(1): 55-65, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24262302

RESUMEN

In its classical form, autophagy is an essential, homeostatic process by which cytoplasmic components are degraded in a double-membrane-bound autophagosome in response to starvation. Paradoxically, although autophagy is primarily a protective process for the cell, it can also play a role in cell death. The roles of autophagy bridge both the innate and adaptive immune systems and autophagic dysfunction is associated with inflammation, infection, neurodegeneration and cancer. In this review, we discuss the contribution of autophagy to inflammatory, infectious and neurodegenerative diseases, as well as cancer.


Asunto(s)
Autofagia , Infecciones/fisiopatología , Neoplasias/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Fagosomas/metabolismo , Inmunidad Adaptativa , Animales , Estructuras Celulares/metabolismo , Homeostasis , Humanos , Inmunidad Innata , Inflamación/fisiopatología
15.
FASEB J ; 27(1): 299-312, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23073832

RESUMEN

Microglial cells are phagocytes in the central nervous system (CNS) and become activated in pathological conditions, resulting in microgliosis, manifested by increased cell numbers and inflammation in the affected regions. Thus, controlling microgliosis is important to prevent pathological damage to the brain. Here, we evaluated the contribution of Toll-like receptor 2 (TLR2) to microglial survival. We observed that activation of microglial cells with peptidoglycan (PGN) from Staphylococcus aureus and other TLR2 ligands results in cell activation followed by the induction of autophagy and autophagy-dependent cell death. In C57BL/6J mice, intracerebral injection of PGN increased the autophagy of microglial cells and reduced the microglial/macrophage cell number in brain parenchyma. Our results demonstrate a novel role of TLRs in the regulation of microglial cell activation and survival, which are important for the control of microgliosis and associated inflammatory responses in the CNS.


Asunto(s)
Autofagia , Muerte Celular/fisiología , Microglía/citología , Polisacáridos/fisiología , Receptor Toll-Like 2/metabolismo , Animales , Western Blotting , Citometría de Flujo , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Transmisión , Polisacáridos/metabolismo
17.
Neurobiol Dis ; 43(3): 616-24, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21624466

RESUMEN

Microglial cells are resident macrophages in the central nervous system (CNS) and become activated in many pathological conditions. Activation of microglial cells results in reactive microgliosis, manifested by an increase in cell number in the affected CNS regions. The control of microgliosis may be important to prevent pathological damage to the brain. The type 2 cytokine IL-4 has been reported to be protective in brain inflammation. However, its effect on microglial cell survival was not well understood. In this study, we report a dual effect of IL-4 on the survival of mouse microglial cells. In a 6h short term culture, IL-4 reduced the death of microglial cells induced by staurosporine. In contrast, in long term treatment (more than 48h), IL-4 increased the apoptotic death of both primary mouse microglial cells and a microglial cell line N9. Mechanistic studies revealed that, in microglial cells, IL-4 increased the levels of cleaved caspase 3 and PARP, which is down-stream of activated caspase 3. In addition, IL-4 down regulated the autophagy and the antiapoptotic protein Bcl-xL in microglial cells. On the other hand, the pre-incubation of microglial cells with IL-4 for 24h, attenuated the cell death induced by the neurotoxic peptide amyloid beta 1-42 (Aß42). Our observations demonstrate a novel function of IL-4 in regulating the survival of microglial cells, which may have important significance in reduction of undesired inflammatory responses in the CNS.


Asunto(s)
Apoptosis/inmunología , Caspasa 3/fisiología , Interleucina-4/fisiología , Microglía/inmunología , Péptidos beta-Amiloides/toxicidad , Animales , Inhibidores de Caspasas , Supervivencia Celular/inmunología , Células Cultivadas , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/enzimología , Microglía/patología , Fragmentos de Péptidos/toxicidad , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Poli(ADP-Ribosa) Polimerasas/metabolismo , Estaurosporina/farmacología
18.
Int Immunopharmacol ; 11(10): 1415-21, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21616174

RESUMEN

The activation of innate immune response is initiated by engagement of pattern-recognition receptors (PPRs), such as Toll-like receptors (TLRs). These receptors are expressed in peripheral leukocytes and in many cell types in the central nervous system (CNS). The expression of TLRs in CNS was mainly studied in astrocytes and microglial cells. However, new evidence indicates that these receptors may play an important role in neuronal homeostasis. The expression of TLRs in the CNS is variable and can be modulated by multiple factors, including pro-inflammatory molecules, which are elevated in neurodegenerative diseases and can increase the expression of TLRs in CNS cells. Moreover, activation of TLRs induces the release of pro-inflammatory cytokines. Therefore, TLRs have been shown to play a role in several aspects of neurodegenerative diseases. Here we will discuss results reported in the recent literature concerning the participation of TLRs in neurodegenerative diseases.


Asunto(s)
Sistema Nervioso Central/metabolismo , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Enfermedades Neurodegenerativas/inmunología , Receptores Toll-Like/inmunología , Animales , Astrocitos/inmunología , Supervivencia Celular/inmunología , Microambiente Celular/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación , Mediadores de Inflamación/inmunología , Microglía/inmunología , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/fisiología , Receptores Toll-Like/agonistas , Receptores Toll-Like/antagonistas & inhibidores
19.
J Immunol ; 183(1): 740-8, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19535628

RESUMEN

IL-12 is an excellent candidate for the treatment of cancer due to its ability to drive strong antitumor responses. Recombinant IL-12 protein is currently used in cancer patients; however, systemic expression of rIL-12 presents disadvantages including cost and dose limitation due to its toxicity. In this study, we used hydrodynamic shear of cDNA as a tool to achieve systemic expression of IL-12. We found that sustained but toxic levels of serum IL-12 could be generated in 6- to 7-wk-old B6 mice after a single injection of the cDNA. Unexpectedly, we observed that when IL-12 cDNA is coinjected with IL-18 cDNA, IL-12 antitumor activity was maintained, but there was a significant attenuation of IL-12 toxicity, as evidenced by a greater survival index and a diminution of liver enzymes (ALT and AST). Interestingly, after IL-12 plus IL-18 cDNA administration, more rapid and higher IL-10 levels were observed than after IL-12 cDNA treatment alone. To understand the mechanism of protection, we coinjected IL-12 plus IL-10 cDNAs and observed an increase in survival that correlated with diminished serum levels of the inflammatory cytokines TNF-alpha and IFN-gamma. Confirming the protective role of early IL-10 expression, we observed a significant decrease in survival in IL-10 knockout mice or IL-10R-blocked B6 mice after IL-12 plus IL-18 treatment. Thus, our data demonstrate that the high and early IL-10 expression induced after IL-12 plus IL-18 cDNA treatment is critical to rapidly attenuate IL-12 toxicity without affecting its antitumor capacity. These data could highly contribute to the design of more efficient/less toxic protocols for the treatment of cancer.


Asunto(s)
Antineoplásicos/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/uso terapéutico , Interleucina-12/biosíntesis , Interleucina-12/toxicidad , Interleucina-18/biosíntesis , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Animales , Antineoplásicos/sangre , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , ADN Complementario/administración & dosificación , ADN Complementario/biosíntesis , Quimioterapia Combinada , Interferón gamma/antagonistas & inhibidores , Interferón gamma/sangre , Interferón gamma/deficiencia , Interleucina-10/deficiencia , Interleucina-12/sangre , Interleucina-12/uso terapéutico , Interleucina-18/fisiología , Interleucina-18/uso terapéutico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/sangre
20.
Int Immunopharmacol ; 7(10): 1271-85, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17673142

RESUMEN

Members of the Toll-like receptor (TLR) family play key roles in both innate and adaptive immune responses. TLR proteins enable host to recognize a large number of pathogen-associated molecular patterns such as bacterial lipopolysaccharides, viral RNA, CPG-containing DNA, and flagellin, among others. TLRs are also apparently able to mediate responses to host molecules, including one defensin, ROS, HMGB1 (high-mobility group box protein 1), surfactant protein A, fibrinogen, breakdown products of tissue matrix, heat shock proteins (hsp) and eosinophil-derived neurotoxin (EDN). Thus, TLR are involved in the development of many pathological conditions including infectious diseases, tissue damage, autoimmune and neurodegenerative diseases and cancer. In this review, the contribution of TLRs to diseases of the central nervous system (CNS), lung, gastrointestinal tract, kidney and skin as well as cancer is evaluated. We hope to provide new insight into the pathogenesis and progression of diseases and more importantly, into the potential for TLRs as targets of therapeutics.


Asunto(s)
Infecciones Bacterianas/inmunología , Inflamación/inmunología , Neoplasias/inmunología , Receptores Toll-Like/inmunología , Virosis/inmunología , Animales , Humanos , Ligandos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...