Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Med Hypotheses ; 1862024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38617026

RESUMEN

Inflamm-aging is a condition of low-grade and chronic systemic inflammation characterized by a systemic increase in multiple inflammatory biomarkers such as tumor necrosis factor (TNF), interleukin 6 (IL-6), C-reactive protein (CRP), and CXCL9 (MIG) in experimental and clinical settings. However, despite the recent identification of extracellular procathepsin L (pCTS-L) as a novel mediator of inflammatory diseases such as sepsis, its possible role in inflamm-aging was previously not investigated. In the present study, we compared blood levels of pCTS-L and other 62 cytokines and chemokines between young and aged Balb/C mice by Western blotting and Cytokine Antibody Arrays. In light of the surprising finding of a marked increase in blood pCTS-L levels in aged mice, we propose that blood pCTS-L levels may serve as another biomarker of inflamm-aging. Given the capacity of pCTS-L in inducing various cytokines (e.g., TNF and IL-6), it will be important to test the hypothetic role of pCTS-L in inflamm-aging under experimental and clinical conditions.

2.
Front Immunol ; 15: 1368448, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38550581

RESUMEN

The pathogenic mechanisms of bacterial infections and resultant sepsis are partly attributed to dysregulated inflammatory responses sustained by some late-acting mediators including the procathepsin-L (pCTS-L). It was entirely unknown whether any compounds of the U.S. Drug Collection could suppress pCTS-L-induced inflammation, and pharmacologically be exploited into possible therapies. Here, we demonstrated that a macrophage cell-based screening of a U.S. Drug Collection of 1360 compounds resulted in the identification of progesterone (PRO) as an inhibitor of pCTS-L-mediated production of several chemokines [e.g., Epithelial Neutrophil-Activating Peptide (ENA-78), Monocyte Chemoattractant Protein-1 (MCP-1) or MCP-3] and cytokines [e.g., Interleukin-10 (IL-10) or Tumor Necrosis Factor (TNF)] in primary human peripheral blood mononuclear cells (PBMCs). In vivo, these PRO-entrapping 2,6-dimethal-ß-cyclodextrin (DM-ß-CD) nanoparticles (containing 1.35 mg/kg PRO and 14.65 mg/kg DM-ß-CD) significantly increased animal survival in both male (from 30% to 70%, n = 20, P = 0.041) and female (from 50% to 80%, n = 30, P = 0.026) mice even when they were initially administered at 24 h post the onset of sepsis. This protective effect was associated with a reduction of sepsis-triggered accumulation of three surrogate biomarkers [e.g., Granulocyte Colony Stimulating Factor (G-CSF) by 40%; Macrophage Inflammatory Protein-2 (MIP-2) by 45%; and Soluble Tumor Necrosis Factor Receptor I (sTNFRI) by 80%]. Surface Plasmon Resonance (SPR) analysis revealed a strong interaction between PRO and pCTS-L (KD = 78.2 ± 33.7 nM), which was paralleled with a positive correlation between serum PRO concentration and serum pCTS-L level (ρ = 0.56, P = 0.0009) or disease severity (Sequential Organ Failure Assessment, SOFA; ρ = 0.64, P = 0.0001) score in septic patients. Our observations support a promising opportunity to explore DM-ß-CD nanoparticles entrapping lipophilic drugs as possible therapies for clinical sepsis.


Asunto(s)
Catepsina L , Precursores Enzimáticos , Sepsis , beta-Ciclodextrinas , Humanos , Masculino , Femenino , Ratones , Animales , Progesterona , Leucocitos Mononucleares
3.
Expert Opin Ther Targets ; 27(7): 575-591, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37477229

RESUMEN

INTRODUCTION: Microbial infections and resultant sepsis are leading causes of death in hospitals, representing approximately 20% of total deaths worldwide. Despite the difficulties in translating experimental insights into effective therapies for often heterogenous patient populations, an improved understanding of the pathogenic mechanisms underlying experimental sepsis is still urgently needed. Sepsis is partly attributable to dysregulated innate immune responses manifested by hyperinflammation and immunosuppression at different stages of microbial infections. AREAS COVERED: Here we review our recent progress in searching for late-acting mediators of experimental sepsis and propose high mobility group box 1 (HMGB1) and procathepsin-L (pCTS-L) as potential therapeutic targets for improving outcomes of lethal sepsis and other infectious diseases. EXPERT OPINION: It will be important to evaluate the efficacy of HMGB1- or pCTS-L-targeting agents for the clinical management of human sepsis and other infectious diseases in future studies.


Asunto(s)
Enfermedades Transmisibles , Proteína HMGB1 , Sepsis , Humanos , Sepsis/tratamiento farmacológico , Inmunidad Innata
4.
Int J Mol Sci ; 24(10)2023 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-37239992

RESUMEN

The pathogenesis of microbial infections and sepsis is partly attributable to dysregulated innate immune responses propagated by late-acting proinflammatory mediators such as procathepsin L (pCTS-L). It was previously not known whether any natural product could inhibit pCTS-L-mediated inflammation or could be strategically developed into a potential sepsis therapy. Here, we report that systemic screening of a NatProduct Collection of 800 natural products led to the identification of a lipophilic sterol, lanosterol (LAN), as a selective inhibitor of pCTS-L-induced production of cytokines [e.g., Tumor Necrosis Factor (TNF) and Interleukin-6 (IL-6)] and chemokines [e.g., Monocyte Chemoattractant Protein-1 (MCP-1) and Epithelial Neutrophil-Activating Peptide (ENA-78)] in innate immune cells. To improve its bioavailability, we generated LAN-carrying liposome nanoparticles and found that these LAN-containing liposomes (LAN-L) similarly inhibited pCTS-L-induced production of several chemokines [e.g., MCP-1, Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted (RANTES) and Macrophage Inflammatory Protein-2 (MIP-2)] in human blood mononuclear cells (PBMCs). In vivo, these LAN-carrying liposomes effectively rescued mice from lethal sepsis even when the first dose was given at 24 h post the onset of this disease. This protection was associated with a significant attenuation of sepsis-induced tissue injury and systemic accumulation of serval surrogate biomarkers [e.g., IL-6, Keratinocyte-derived Chemokine (KC), and Soluble Tumor Necrosis Factor Receptor I (sTNFRI)]. These findings support an exciting possibility to develop liposome nanoparticles carrying anti-inflammatory sterols as potential therapies for human sepsis and other inflammatory diseases.


Asunto(s)
Liposomas , Sepsis , Ratones , Humanos , Animales , Liposomas/uso terapéutico , Lanosterol/uso terapéutico , Interleucina-6 , Citocinas , Quimiocinas , Sepsis/patología
5.
Sci Adv ; 9(5): eadf4313, 2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-36735789

RESUMEN

Antibody-based strategies have been attempted to antagonize early cytokines of sepsis, but not yet been tried to target inducible late-acting mediators. Here, we report that the expression and secretion of procathepsin-L (pCTS-L) was induced by serum amyloid A (SAA) in innate immune cells, contributing to its late and systemic accumulation in experimental and clinical sepsis. Recombinant pCTS-L induced interleukin-6 (IL-6), IL-8, GRO-α/KC, GRO-ß/MIP-2, and MCP-1 release in innate immune cells and moderately correlated with blood concentrations of these cytokines/chemokines in clinical sepsis. Mechanistically, pCTS-L interacted with Toll-like receptor 4 (TLR4) and the receptor for advanced glycation end products (RAGE) to induce cytokines/chemokines. Pharmacological suppression of pCTS-L with neutralizing polyclonal and monoclonal antibodies attenuated pCTS-L-mediated inflammation by impairing its interaction with TLR4 and RAGE receptors, and consequently rescued animals from lethal sepsis. Our findings have suggested a possibility of developing antibody strategies to prevent dysregulated immune responses mediated by late-acting cytokines.


Asunto(s)
Sepsis , Receptor Toll-Like 4 , Animales , Receptor Toll-Like 4/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Citocinas , Receptor para Productos Finales de Glicación Avanzada , Quimiocinas/metabolismo
6.
J Leukoc Biol ; 111(1): 261-267, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33759207

RESUMEN

A severe acute respiratory syndrome (SARS)-like coronavirus 2 (SARS-CoV-2) has recently caused a pandemic COVID-19 disease that infected approximately 94 million and killed more than 2,000,000 people worldwide. Like the SARS-CoV, SARS-CoV-2 also employs a receptor-binding motif (RBM) of its envelope spike protein for binding the host angiotensin-converting enzyme 2 (ACE2) to gain viral entry. Currently, extensive efforts are being made to produce vaccines against a surface fragment of a SARS-CoV-2, such as the spike protein, in order to boost protective antibodies that can inhibit virus-ACE2 interaction to prevent viral entry. It was previously unknown how spike protein-targeting antibodies would affect innate inflammatory responses to SARS-CoV-2 infections. Here we generated a highly purified recombinant protein corresponding to the RBM of SARS-CoV-2, and used it to screen for cross-reactive monoclonal antibodies (mAbs). We found two RBM-binding mAbs that competitively inhibited its interaction with human ACE2, and specifically blocked the RBM-induced GM-CSF secretion in both human peripheral blood mononuclear cells and murine macrophage cultures. Our findings have suggested a possible strategy to prevent SARS-CoV-2-elicited "cytokine storm," and revealed a potentially anti-inflammatory and protective mechanism for SARS-CoV-2 spike-based vaccines.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Secuencias de Aminoácidos , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Ratones , Unión Proteica , Células RAW 264.7 , Proteínas Recombinantes/metabolismo
7.
Cells ; 10(9)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34571869

RESUMEN

Sepsis remains a common cause of death in intensive care units, accounting for approximately 20% of total deaths worldwide. Its pathogenesis is partly attributable to dysregulated inflammatory responses to bacterial endotoxins (such as lipopolysaccharide, LPS), which stimulate innate immune cells to sequentially release early cytokines (such as tumor necrosis factor (TNF) and interferons (IFNs)) and late mediators (such as high-mobility group box 1, HMGB1). Despite difficulties in translating mechanistic insights into effective therapies, an improved understanding of the complex mechanisms underlying the pathogenesis of sepsis is still urgently needed. Here, we review recent progress in elucidating the intricate mechanisms underlying the regulation of HMGB1 release and action, and propose a few potential therapeutic candidates for future clinical investigations.


Asunto(s)
Citocinas/inmunología , Proteína HMGB1/inmunología , Lipopolisacáridos/inmunología , Sepsis/inmunología , Animales , Humanos
8.
Front Immunol ; 12: 821154, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095926

RESUMEN

Background: Hepatic ischemia and reperfusion (I/R) injury is commonly associated with surgical liver resection or transplantation, and represents a major cause of liver damage and graft failure. Currently, there are no effective therapies to prevent hepatic I/R injury other than ischemic preconditioning and some preventative strategies. Previously, we have revealed the anti-inflammatory activity of a sweat gland-derived peptide, dermcidin (DCD), in macrophage/monocyte cultures. Here, we sought to explore its therapeutic potential and protective mechanisms in a murine model of hepatic I/R. Methods: Male C57BL/6 mice were subjected to hepatic ischemia by clamping the hepatic artery and portal vein for 60 min, which was then removed to initiate reperfusion. At the beginning of reperfusion, 0.2 ml saline control or solution of DCD (0.5 mg/kg BW) or DCD-C34S analog (0.25 or 0.5 mg/kg BW) containing a Cys (C)→Ser (S) substitution at residue 34 was injected via the internal jugular vein. For survival experiments, mice were subjected to additional resection to remove non-ischemic liver lobes, and animal survival was monitored for 10 days. For mechanistic studies, blood and tissue samples were collected at 24 h after the onset of reperfusion, and subjected to measurements of various markers of inflammation and tissue injury by real-time RT-PCR, immunoassays, and histological analysis. Results: Recombinant DCD or DCD-C34S analog conferred a significant protection against lethal hepatic I/R when given intravenously at the beginning of reperfusion. This protection was associated with a significant reduction in hepatic injury, neutrophilic CXC chemokine (Mip-2) expression, neutrophil infiltration, and associated inflammation. Furthermore, the administration of DCD also resulted in a significant attenuation of remote lung inflammatory injury. Mechanistically, DCD interacted with epidermal growth factor receptor (EGFR), a key regulator of liver inflammation, and significantly inhibited hepatic I/R-induced phosphorylation of EGFR as well as a downstream signaling molecule, protein kinase B (AKT). The suppression of EGFR expression by transducing Egfr-specific shRNA plasmid into macrophages abrogated the DCD-mediated inhibition of nitric oxide (NO) production induced by a damage-associated molecular pattern (DAMP), cold-inducible RNA-binding protein, CIRP. Conclusions: The present study suggests that human DCD and its analog may be developed as novel therapeutics to attenuate hepatic I/R-induced inflammatory injury possibly by impairing EGFR signaling.


Asunto(s)
Antiinflamatorios/farmacología , Dermcidinas/farmacología , Inflamación/etiología , Inflamación/patología , Hepatopatías/complicaciones , Sustancias Protectoras/farmacología , Daño por Reperfusión/complicaciones , Secuencia de Aminoácidos , Animales , Antiinflamatorios/química , Biomarcadores , Biopsia , Citocinas/genética , Citocinas/metabolismo , Dermcidinas/química , Susceptibilidad a Enfermedades , Receptores ErbB/metabolismo , Humanos , Inmunohistoquímica , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Hepatopatías/tratamiento farmacológico , Hepatopatías/etiología , Masculino , Ratones , Infiltración Neutrófila , Óxido Nítrico/metabolismo , Especificidad de Órganos , Fosforilación , Sustancias Protectoras/química , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/etiología
9.
bioRxiv ; 2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32908979

RESUMEN

A severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) has recently caused a pandemic COVID-19 disease that infected more than 25.6 million and killed 852,000 people worldwide. Like the SARS-CoV, SARS-CoV-2 also employs a receptor-binding motif (RBM) of its envelope spike protein for binding the host angiotensin-converting enzyme 2 (ACE2) to gain viral entry. Currently, extensive efforts are being made to produce vaccines against a surface fragment of a SARS-CoV-2, such as the spike protein, in order to boost protective antibody responses. It was previously unknown how spike protein-targeting antibodies would affect innate inflammatory responses to SARS-CoV-2 infections. Here we generated a highly purified recombinant protein corresponding to the RBM of SARS-CoV-2, and used it to screen for cross-reactive monoclonal antibodies (mAbs). We found two RBM-binding mAbs that competitively inhibited its interaction with human ACE2, and specifically blocked the RBM-induced GM-CSF secretion in both human monocyte and murine macrophage cultures. Our findings have suggested a possible strategy to prevent SARS-CoV-2-elicited "cytokine storm", and provided a potentially useful criteria for future assessment of innate immune-modulating properties of various SARS-CoV-2 vaccines. ONE SENTENCE SUMMARY: RBM-binding Antibodies Inhibit GM-CSF Induction.

10.
Sci Transl Med ; 12(539)2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32295901

RESUMEN

For the clinical management of sepsis, antibody-based strategies have only been attempted to antagonize proinflammatory cytokines but not yet been tried to target harmless proteins that may interact with these pathogenic mediators. Here, we report an antibody strategy to intervene in the harmful interaction between tetranectin (TN) and a late-acting sepsis mediator, high-mobility group box 1 (HMGB1), in preclinical settings. We found that TN could bind HMGB1 to reciprocally enhance their endocytosis, thereby inducing macrophage pyroptosis and consequent release of lactate dehydrogenase and apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain. The genetic depletion of TN expression or supplementation of exogenous TN protein at subphysiological doses distinctly affected the outcomes of potentially lethal sepsis, revealing a previously underappreciated beneficial role of TN in sepsis. Furthermore, the administration of domain-specific polyclonal and monoclonal antibodies effectively inhibited TN/HMGB1 interaction and endocytosis and attenuated the sepsis-induced TN depletion and tissue injury, thereby rescuing animals from lethal sepsis. Our findings point to a possibility of developing antibody strategies to prevent harmful interactions between harmless proteins and pathogenic mediators of human diseases.


Asunto(s)
Proteína HMGB1 , Lectinas Tipo C , Sepsis , Animales , Anticuerpos Monoclonales/uso terapéutico , Humanos , Sepsis/tratamiento farmacológico
12.
J Affect Disord ; 259: 56-66, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31437702

RESUMEN

BACKGROUND: Associations between abnormal gut microbiome compositions and anxiety-like behaviors are well established. However, it is unknown whether the gut microbiome composition is associated with the severity of generalized anxiety disorder (GAD) and relief from clinical symptoms in patients. METHODS: Stool samples from 36 patients with active GAD (A-GAD group) and 24 matched healthy control subjects (HC group) were analyzed by 16S rRNA gene sequencing. Anxiety was assessed with the Hamilton Anxiety Rating Scale and the Self-rating Anxiety Scale, and global assessments of functioning were performed at baseline and 1 month after drug treatment. RESULTS: Gut microbiome compositions were altered in A-GAD patients, with fewer operational taxonomic units and lower fecal bacterial α-diversity. Specifically, Firmicutes and Tenericutes abundances were lower in A-GAD patients, and several genera were differentially represented in the A-GAD and HC groups. The abundances of Eubacterium_coprostanoligenes_group, Ruminococcaceae_UCG-014, and Prevotella_9 correlated negatively with the anxiety severity and positively with anxiety reduction, whereas the abundances of Bacteroides and Escherichia-Shigella were positively associated with anxiety severity. Sex, smoking, and alcohol intake influenced the gut microbiome composition. LIMITATIONS: The sample sizes were small and the stool samples were collected only at baseline; therefore, a causal association between changes in intestinal flora and disease remission was not established. Moreover, the effects of different drugs on gut microbiome composition were not investigated. CONCLUSIONS: Altered gut microbiome composition may contribute to GAD pathogenesis and remission.


Asunto(s)
Trastornos de Ansiedad/microbiología , Heces/microbiología , Microbiota , ARN Ribosómico 16S/análisis , Adulto , Femenino , Microbioma Gastrointestinal , Humanos , Masculino
13.
Sci Rep ; 9(1): 160, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30655582

RESUMEN

We have recently reported an important role of Connexin 43 (Cx43) hemichannels in the pathogenesis of lethal sepsis through facilitating ATP efflux to potentiate the double-stranded RNA-activated protein kinase R (PKR)-dependent macrophage activation. Here we further elucidated the possible role of Pannexin 1 (Panx1) hemichannel in lethal sepsis by assessing its expression along with the impact of a Panx1-specific mimetic inhibitory peptide, 10Panx, on macrophage hemichannel activity in vitro and animal sepsis lethality in vivo. Both crude bacterial lipopolysaccharide (LPS) and purified serum amyloid A (SAA) effectively induced the expression and extracellular release of Panx1 by macrophages or monocytes as judged by Western blotting and immunocytochemistry assays. In animal model of lethal sepsis, Panx1 expression levels were significantly elevated in the heart, but reduced in the kidney, lung, spleen, and blood. At relatively lower doses (10, 50, and 100 mg/kg), the Panx1 mimetic peptide, 10Panx, reproducibly exacerbated the sepsis-induced animal lethality, reducing survival rates from 60-70% to 0-10%. Consistently, 10Panx did not inhibit, but rather promoted, the LPS-induced elevation of Lucifer Yellow dye uptake, ATP release, and Nitric Oxide (NO) production. Collectively, these findings suggested that elevated macrophage Panx1 expression and hemichannel activation contribute to the pathogenesis of lethal sepsis.


Asunto(s)
Conexinas/fisiología , Proteínas del Tejido Nervioso/fisiología , Sepsis/metabolismo , Animales , Células Cultivadas , Conexinas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Macrófagos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/antagonistas & inhibidores
14.
Sci Rep ; 8(1): 166, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317708

RESUMEN

Cytoplasmic membrane-bound connexin 43 (Cx43) proteins oligomerize into hexameric channels (hemichannels) that can sometimes dock with hemichannels on adjacent cells to form gap junctional (GJ) channels. However, the possible role of Cx43 hemichannels in sterile and infectious inflammatory diseases has not been adequately defined due to the lack of selective interventions. Here we report that a proinflammatory mediator, the serum amyloid A (SAA), resembled bacterial endotoxin by stimulating macrophages to up-regulate Cx43 expression and double-stranded RNA-activated protein kinase R (PKR) phosphorylation in a TLR4-dependent fashion. Two well-known Cx43 mimetic peptides, the GAP26 and TAT-GAP19, divergently affected macrophage hemichannel activities in vitro, and differentially altered the outcome of lethal sepsis in vivo. By screening a panel of Cx43 mimetic peptides, we discovered that one cysteine-containing peptide, P5 (ENVCYD), effectively attenuated hemichannel activities, and significantly suppressed endotoxin-induced release of ATP and HMGB1 in vitro. In vivo, the P5 peptide conferred a significant protection against hepatic ischemia/reperfusion injury and lethal microbial infection. Collectively, these findings have suggested a pathogenic role of Cx43 hemichannels in sterile injurious as well as infectious inflammatory diseases possibly through facilitating extracellular ATP efflux to trigger PKR phosphorylation/activation.


Asunto(s)
Conexina 43/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Conexina 43/antagonistas & inhibidores , Conexina 43/química , Endotoxinas/metabolismo , Humanos , Inflamación/etiología , Inflamación/mortalidad , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Péptidos/metabolismo , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Sepsis/etiología , Sepsis/metabolismo , Sepsis/mortalidad , eIF-2 Quinasa/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-29152323

RESUMEN

E. coli releases a 33 amino acid peptide melanocortin-like peptide of E. coli (MECO-1) that is identical to the C-terminus of the E. coli elongation factor-G (EF-G) and has interesting similarities to two prominent mammalian melanocortin hormones, alpha-melanocyte-stimulating hormone (alpha-MSH) and adrenocorticotropin (ACTH). Note that MECO-1 lacks HFRW, the common pharmacophore of the known mammalian melanocortin peptides. MECO-1 and the two hormones were equally effective in severely blunting release of cytokines (HMGB1 and TNF) from macrophage-like cells in response to (i) endotoxin (lipopolysaccharide) or (ii) pro-inflammatory cytokine HMGB-1. The in vitro anti-inflammatoty effects of MECO-1 and of alpha-MSH were abrogated by (i) antibody against melanocortin-1 receptor (MC1R) and by (ii) agouti, an endogenous inverse agonist of MC1R. In vivo MECO-1 was even more potent than alpha-MSH in rescuing mice from death due to (i) lethal doses of LPS endotoxin or (ii) cecal ligation and puncture, models of sterile and infectious sepsis, respectively.

16.
Shock ; 45(1): 28-32, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26529659

RESUMEN

The epidermal barriers of the skin serve as the first layer of defense by limiting the access of many pathogens to the blood circulation. In addition, human skin also contains sweat glands that can secrete a wide array of antimicrobial peptides to restrain the growth of various microbes. In the case of microbial infection, macrophages and monocytes constitute the first line of defense by producing a wide array of proinflammatory cytokines and chemokines. This process is triggered either by pathogen-associated molecular pattern molecules (such as bacterial endotoxin) or damage-associated molecular pattern molecules (such as HMGB1). In light of our findings that a sweat gland-derived antimicrobial peptide, dermcidin, affected both pathogen-associated molecular pattern and damage-associated molecular pattern-induced cytokines/chemokines by macrophages/monocytes, we propose that dermcidin may play an important role in the regulation of the innate immune responses to infection and injury. Future investigations are warranted to further test this understudied hypothesis in both preclinical and clinical settings.


Asunto(s)
Antiinfecciosos/inmunología , Dermcidinas/inmunología , Secuencia de Aminoácidos , Animales , Antiinfecciosos/administración & dosificación , Antiinfecciosos/química , Células Cultivadas , Quimiocinas/biosíntesis , Dermcidinas/administración & dosificación , Dermcidinas/química , Relación Dosis-Respuesta Inmunológica , Proteína HMGB1/inmunología , Humanos , Inmunidad Innata/inmunología , Lipopolisacáridos/inmunología , Ratones , Datos de Secuencia Molecular , Profármacos/química , Células RAW 264.7 , Glándulas Sudoríparas/química
17.
Biochim Biophys Acta ; 1840(7): 2253-61, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24613680

RESUMEN

BACKGROUND: Neuroinflammation is a key cascade after cerebral ischemia. Excessive production of proinflammatory mediators in ischemia exacerbates brain injury. Cold-inducible RNA-binding protein (CIRP) is a newly discovered proinflammatory mediator that can be released into the circulation during hemorrhage or septic shock. Here, we examine the involvement of CIRP in brain injury during ischemic stroke. METHODS: Stroke was induced by middle cerebral artery occlusion (MCAO). In vitro hypoxia was conducted in a hypoxia chamber containing 1% oxygen. CIRP and tumor necrosis factor-α (TNF-α) levels were assessed by RT-PCR and Western blot analysis. RESULTS: CIRP is elevated along with an upregulation of TNF-α expression in mouse brain after MCAO. In CIRP-deficient mice, the brain infarct volume, induction of TNF-α, and activation of microglia are markedly reduced after MCAO. Using microglial BV2 cells, we demonstrate that hypoxia induces the expression, translocation, and release of CIRP, which is associated with an increase of TNF-α levels. Addition of recombinant murine (rm) CIRP directly induces TNF-α release from BV2 cells and such induction is inhibited by neutralizing antisera to CIRP. Moreover, rmCIRP activates the NF-κB signaling pathway in BV2 cells. The conditioned medium from BV2 cells exposed to hypoxia triggers the apoptotic cascade by increasing caspase activity and decreasing Bcl-2 expression in neural SH-SY5Y cells, which is inhibited by antisera to CIRP. CONCLUSION: Extracellular CIRP is a detrimental factor in stimulating inflammation to cause neuronal damage in cerebral ischemia. GENERAL SIGNIFICANCE: Development of an anti-CIRP therapy may benefit patients with brain ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Neuronas/patología , Proteínas de Unión al ARN/metabolismo , Accidente Cerebrovascular/patología , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Infarto de la Arteria Cerebral Media , Inflamación/complicaciones , Inflamación/metabolismo , Inflamación/patología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Neuronas/metabolismo , Proteínas de Unión al ARN/genética , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
18.
Nat Med ; 19(11): 1489-1495, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24097189

RESUMEN

A systemic inflammatory response is observed in patients undergoing hemorrhagic shock and sepsis. Here we report increased levels of cold-inducible RNA-binding protein (CIRP) in the blood of individuals admitted to the surgical intensive care unit with hemorrhagic shock. In animal models of hemorrhage and sepsis, CIRP is upregulated in the heart and liver and released into the circulation. In macrophages under hypoxic stress, CIRP translocates from the nucleus to the cytosol and is released. Recombinant CIRP stimulates the release of tumor necrosis factor-α (TNF-α) and HMGB1 from macrophages and induces inflammatory responses and causes tissue injury when injected in vivo. Hemorrhage-induced TNF-α and HMGB1 release and lethality were reduced in CIRP-deficient mice. Blockade of CIRP using antisera to CIRP attenuated inflammatory cytokine release and mortality after hemorrhage and sepsis. The activity of extracellular CIRP is mediated through the Toll-like receptor 4 (TLR4)-myeloid differentiation factor 2 (MD2) complex. Surface plasmon resonance analysis indicated that CIRP binds to the TLR4-MD2 complex, as well as to TLR4 and MD2 individually. In particular, human CIRP amino acid residues 106-125 bind to MD2 with high affinity. Thus, CIRP is a damage-associated molecular pattern molecule that promotes inflammatory responses in shock and sepsis.


Asunto(s)
Proteínas y Péptidos de Choque por Frío/sangre , Proteínas de Unión al ARN/sangre , Sepsis/sangre , Choque Hemorrágico/sangre , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Neutralizantes/administración & dosificación , Hipoxia de la Célula , Línea Celular , Proteínas y Péptidos de Choque por Frío/antagonistas & inhibidores , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/sangre , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba
19.
Int J Mol Med ; 28(6): 1071-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21874225

RESUMEN

Apoptosis plays an important role in the patho-biology of sepsis. The opsonizing protein milk fat globule-EGF factor VIII (MFG-E8) is involved in apoptotic cell clearance. Our previous studies have shown that administration of rat MFG-E8-containing exosomes or recombinant murine MFG-E8 (rmMFG-E8) is protective in a rat model of sepsis induced by cecal ligation of puncture (CLP). However, one obstacle hampering the development of MFG-E8 as a therapeutic agent for septic patients is the potential immunogenicity of animal proteins in humans. The purpose of this study, therefore, was to express recombinant human MFG-E8 (rhMFG-E8) and characterize its biological activity. Using an E. coli system, we successfully expressed and purified the mature molecule of human MFG-E8 (Leu24-Cys387). The purity of rhMFG-E8 was over 99% and it was immunoreactive for specific anti-human MFG-E8 antibodies. Amino acid sequence analysis by LC-MS/MS identified the purified protein as human MFG-E8. Using primary rat peritoneal macrophages, we showed that rhMFG-E8 markedly increased peritoneal macrophage phagocytosis of apoptotic thymocytes, which was as effective as commercial rmMFG-E8. To determine the biological activity of rhMFG-E8 in vivo, male adult rats were subjected to sepsis by CLP. rhMFG-E8 or rmMFG-E8 were administered intravenously at the time of CLP. Our results demonstrated that both rhMFG-E8 and rmMFG-E8 reduced thymocyte apoptosis and plasma levels of lactate and IL-6 at 20 h after CLP, and improved the 10-day survival rate. Thus, we have successfully expressed and purified biologically active rhMFG-E8. Our newly-expressed rhMFG-E8 is highly effective in the rat model of sepsis.


Asunto(s)
Antígenos de Superficie/uso terapéutico , Macrófagos Peritoneales/efectos de los fármacos , Proteínas de la Leche/uso terapéutico , Fagocitosis/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico , Sepsis/tratamiento farmacológico , Animales , Antígenos de Superficie/administración & dosificación , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/genética , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Escherichia coli , Expresión Génica , Glucolípidos/química , Glicoproteínas/química , Humanos , Inyecciones Intravenosas , Interleucina-6/análisis , Interleucina-6/biosíntesis , Gotas Lipídicas , Masculino , Ratones , Proteínas de la Leche/administración & dosificación , Proteínas de la Leche/biosíntesis , Proteínas de la Leche/genética , Leche Humana/química , Plásmidos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Sepsis/inmunología , Sepsis/patología , Timocitos/citología , Transformación Bacteriana
20.
Mol Med ; 17(9-10): 1075-83, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695352

RESUMEN

Stroke is a leading cause of death and the primary medical cause of acquired adult disability worldwide. The progressive brain injury after acute stroke is partly mediated by ischemia-elicited inflammatory responses. The vasoactive hormone adrenomedullin (AM), upregulated under various inflammatory conditions, counterbalances inflammatory responses. However, regulation of AM activity in ischemic stroke remains largely unknown. Recent studies have demonstrated the presence of a specific AM binding protein (that is, AMBP-1) in mammalian blood. AMBP-1 potentiates AM biological activities. Using a rat model of focal cerebral ischemia induced by permanent middle cerebral artery occlusion (MCAO), we found that plasma levels of AM increased significantly, whereas plasma levels of AMBP-1 decreased significantly after stroke. When given peripherally early after MCAO, exogenous human AM in combination with human AMBP-1 reduced brain infarct volume 24 and 72 h after MCAO, an effect not observed after the treatment by human AM or human AMBP-1 alone. Furthermore, treatment of human AM/AMBP-1 reduced neuron apoptosis and morphological damage, inhibited neutrophil infiltration in the brain and decreased serum levels of S100B and lactate. Thus, human AM/AMBP-1 has the ability to reduce stroke-induced brain injury in rats. AM/AMBP-1 can be developed as a novel therapeutic agent for patients with ischemic stroke.


Asunto(s)
Adrenomedulina/farmacología , Apoptosis/efectos de los fármacos , Lesiones Encefálicas/prevención & control , Factor H de Complemento/farmacología , Adrenomedulina/sangre , Adrenomedulina/genética , Animales , Western Blotting , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/complicaciones , Cardiotónicos/sangre , Cardiotónicos/metabolismo , Cardiotónicos/farmacología , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Sinergismo Farmacológico , Humanos , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/etiología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/prevención & control , Lactatos/metabolismo , Masculino , Factores de Crecimiento Nervioso/metabolismo , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/metabolismo , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA