Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(3): 113942, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38489266

RESUMEN

Tumor-associated macrophages (TAMs) shape tumor immunity and therapeutic efficacy. However, it is poorly understood whether and how post-translational modifications (PTMs) intrinsically affect the phenotype and function of TAMs. Here, we reveal that peptidylarginine deiminase 4 (PAD4) exhibits the highest expression among common PTM enzymes in TAMs and negatively correlates with the clinical response to immune checkpoint blockade. Genetic and pharmacological inhibition of PAD4 in macrophages prevents tumor progression in tumor-bearing mouse models, accompanied by an increase in macrophage major histocompatibility complex (MHC) class II expression and T cell effector function. Mechanistically, PAD4 citrullinates STAT1 at arginine 121, thereby promoting the interaction between STAT1 and protein inhibitor of activated STAT1 (PIAS1), and the loss of PAD4 abolishes this interaction, ablating the inhibitory role of PIAS1 in the expression of MHC class II machinery in macrophages and enhancing T cell activation. Thus, the PAD4-STAT1-PIAS1 axis is an immune restriction mechanism in macrophages and may serve as a cancer immunotherapy target.


Asunto(s)
Hidrolasas , Procesamiento Proteico-Postraduccional , Ratones , Animales , Desiminasas de la Arginina Proteica/metabolismo , Arginina Deiminasa Proteína-Tipo 4/genética , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Hidrolasas/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Macrófagos/metabolismo
2.
Clin Rheumatol ; 43(1): 129-135, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37792147

RESUMEN

OBJECTIVE: This study aimed to assess the role of synovial fluid (SF) CD4+T, CD19+B, follicular helper cells (Tfh), and cytokines in the pathogenesis of rheumatoid arthritis (RA). METHODS: This study enrolled 16 patients with RA and 8 patients with osteoarthritis (OA). The frequencies of the SF CD4+ T, CD19+ B, Tfh cells, and Tfh subsets were assessed using flow cytometry. The medical condition in patients with RA was evaluated using The Disease Activity Score 28 (DAS28), the Clinical Disease Activity Index (CDAI), and the Simplified Disease Activity Index (SDAI). Levels of C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), anti-cyclic citrullinated peptide (anti-CCP), and rheumatoid factor (RF) were measured. The cytokines IL-4, IL-13, IL-21, and BLyS were measured by ELISA test. RESULTS: The percentages of SF CD4+T, CD19+B, and PD-1+CXCR5+ Tfh in RA patients were higher than those in OA patients. And the Tfh2 was the main subset among Tfh subsets. In addition, levels of IL-21 and BLyS were higher in patients with RA compared to patients with OA. Furthermore, the treatment of TNF-α inhibitors may be associated with decreased levels of SF Tfh. CONCLUSIONS: Elevated SF Tfh, B cell, and cytokines expression profiles were observed in RA patients. Tfh2 was the major subset of the Tfh, and IL-21 and BLyS were significantly enhanced. Additionally, TNF-α inhibitors reduced Tfh in SF. Therefore, Tfh, B, and Tfh2 cells could play a significant role in the progression of RA. Key Points •Tfh cells in the synovial fluid are significantly higher in RA patients and are dominated by the Tfh2 subpopulation. •Synovial fluid Tfh cells decrease in RA patients after anti-TNF-α treatment.


Asunto(s)
Artritis Reumatoide , Osteoartritis , Humanos , Citocinas , Células T Auxiliares Foliculares/metabolismo , Líquido Sinovial/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Inhibidores del Factor de Necrosis Tumoral , Linfocitos T Colaboradores-Inductores , Osteoartritis/metabolismo
3.
Front Immunol ; 12: 761345, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950139

RESUMEN

Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to infection that often results in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). An emerging mechanism of sepsis-induced ARDS involves neutrophils/macrophages undergoing cell death, releasing nuclear histones to cause tissue damage that exacerbates pulmonary injury. While published studies focus on unmodified histones, little is known about the role of citrullinated histone H3 (CitH3) in the pathogenesis of sepsis and ALI. In this study, we found that levels of CitH3 were elevated in the patients with sepsis-induced ARDS and correlated to PaO2/FiO2 in septic patients. Systematic administration of CitH3 peptide in mice provoked Caspase-1 activation in the lung tissue and caused ALI. Neutralization of CitH3 with monoclonal antibody improved survival and attenuated ALI in a mouse sepsis model. Furthermore, we demonstrated that CitH3 induces ALI through activating Caspase-1 dependent inflammasome in bone marrow derived macrophages and bone marrow derived dendritic cells. Our study suggests that CitH3 is an important mediator of inflammation and mortality during sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Histonas/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Sepsis/inmunología , Lesión Pulmonar Aguda/etiología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Caspasa 1/inmunología , Células Cultivadas , Citrulinación , Células Dendríticas/inmunología , Humanos , Inflamasomas/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Péptidos/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Síndrome de Dificultad Respiratoria/etiología , Sepsis/complicaciones
4.
Front Immunol ; 12: 761946, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804050

RESUMEN

Peptidylarginine deiminases (PADs) are a group of enzymes that catalyze post-translational modifications of proteins by converting arginine residues into citrullines. Among the five members of the PAD family, PAD2 and PAD4 are the most frequently studied because of their abundant expression in immune cells. An increasing number of studies have identified PAD2 as an essential factor in the pathogenesis of many diseases. The successes of preclinical research targeting PAD2 highlights the therapeutic potential of PAD2 inhibition, particularly in sepsis and autoimmune diseases. However, the underlying mechanisms by which PAD2 mediates host immunity remain largely unknown. In this review, we will discuss the role of PAD2 in different types of cell death signaling pathways and the related immune disorders contrasted with functions of PAD4, providing novel therapeutic strategies for PAD2-associated pathology.


Asunto(s)
Arginina Deiminasa Proteína-Tipo 2/inmunología , Animales , Humanos , Enfermedades del Sistema Inmune/inmunología , Infecciones/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Neoplasias/inmunología
5.
Surg Infect (Larchmt) ; 22(4): 421-426, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32833601

RESUMEN

Objective: Sepsis causes millions of deaths on a global scale annually. Activation of peptidylarginine deiminase (PAD) enzymes in sepsis causes citrullination of histones, which results in neutrophil extracellular trap formation and sepsis progression. This study evaluates pan-PAD inhibitor, Cl-amidine, in a model of lipopolysaccharide (LPS)-induced endotoxic shock in rabbits. We hypothesized that Cl-amidine would improve survival and attenuate kidney injury. Methods: In the survival model, rabbits were injected injected intravenously with 1 mg/kg of LPS, and then randomly assigned either to receive dimethyl sulfoxide (DMSO; 1 mcL/g) or Cl-amidine (10 mg/kg diluted in 1 mcL/g DMSO). They were then monitored for 14 days to evaluate survival. In the non-survival experiment, the same insult and treatment were administered, however; the animals were euthanized 12 hours after LPS injection for kidney harvest. Acute kidney injury (AKI) scoring was performed by a histopathologist who was blinded to the group assignment. Serial blood samples were also collected and compared. Results: Rabbits that received Cl-amidine had a higher survival (72%) compared with the rabbits that received DMSO (14%; p < 0.05). Cl-amidine-treated rabbits had lower (p < 0.05) histopathologic AKI scores, as well as plasma creatinine and blood urea nitrogen (BUN) levels 12 hours after insult. Conclusions: Pan-PAD inhibitor Cl-amidine improves survival and attenuates kidney injury in LPS-induced endotoxic shock in rabbits.


Asunto(s)
Trampas Extracelulares , Choque Séptico , Animales , Conejos , Riñón , Lipopolisacáridos/toxicidad , Ornitina/análogos & derivados , Choque Séptico/tratamiento farmacológico
6.
Blood ; 137(12): 1591-1602, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33275650

RESUMEN

Digital protein assays have great potential to advance immunodiagnostics because of their single-molecule sensitivity, high precision, and robust measurements. However, translating digital protein assays to acute clinical care has been challenging because it requires deployment of these assays with a rapid turnaround. Herein, we present a technology platform for ultrafast digital protein biomarker detection by using single-molecule counting of immune-complex formation events at an early, pre-equilibrium state. This method, which we term "pre-equilibrium digital enzyme-linked immunosorbent assay" (PEdELISA), can quantify a multiplexed panel of protein biomarkers in 10 µL of serum within an unprecedented assay incubation time of 15 to 300 seconds over a 104 dynamic range. PEdELISA allowed us to perform rapid monitoring of protein biomarkers in patients manifesting post-chimeric antigen receptor T-cell therapy cytokine release syndrome, with ∼30-minute sample-to-answer time and a sub-picograms per mL limit of detection. The rapid, sensitive, and low-input volume biomarker quantification enabled by PEdELISA is broadly applicable to timely monitoring of acute disease, potentially enabling more personalized treatment.


Asunto(s)
Citocinas/sangre , Enfermedades del Sistema Inmune/sangre , Pruebas en el Punto de Atención , Biomarcadores/sangre , Proteínas Sanguíneas/análisis , Ensayo de Inmunoadsorción Enzimática , Diseño de Equipo , Humanos
7.
Infection ; 49(1): 83-93, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33000445

RESUMEN

PURPOSE: Microbial infection stimulates neutrophil/macrophage/monocyte extracellular trap formation, which leads to the release of citrullinated histone H3 (CitH3) catalyzed by peptidylarginine deiminase (PAD) 2 and 4. Understanding these molecular mechanisms in the pathogenesis of septic shock will be an important next step for developing novel diagnostic and treatment modalities. We sought to determine the expression of CitH3 in patients with septic shock, and to correlate CitH3 levels with PAD2/PAD4 and clinically relevant outcomes. METHODS: Levels of CitH3 were measured in serum samples of 160 critically ill patients with septic and non-septic shock, and healthy volunteers. Analyses of clinical and laboratory characteristics of patients were conducted. RESULTS: Levels of circulating CitH3 at enrollment were significantly increased in septic shock patients (n = 102) compared to patients hospitalized with non-infectious shock (NIC) (n = 32, p < 0.0001). The area under the curve (95% CI) for distinguishing septic shock from NIC using CitH3 was 0.76 (0.65-0.86). CitH3 was positively correlated with PAD2 and PAD4 concentrations and Sequential Organ Failure Assessment Scores [total score (r = 0.36, p < 0.0001)]. The serum levels of CitH3 at 24 h (p < 0.01) and 48 h (p < 0.05) were significantly higher in the septic patients that did not survive. CONCLUSION: CitH3 is increased in patients with septic shock. Its serum concentrations correlate with disease severity and prognosis, which may yield vital insights into the pathophysiology of sepsis.


Asunto(s)
Citrulina/metabolismo , Histonas , Choque Séptico/diagnóstico , Choque/diagnóstico , Anciano , Diagnóstico Diferencial , Femenino , Histonas/sangre , Histonas/química , Humanos , Masculino , Persona de Mediana Edad , Polipéptido alfa Relacionado con Calcitonina/sangre , Arginina Deiminasa Proteína-Tipo 2/sangre , Arginina Deiminasa Proteína-Tipo 4/sangre , Estudios Retrospectivos , Choque/sangre , Choque/epidemiología , Choque Séptico/sangre , Choque Séptico/epidemiología , Resultado del Tratamiento
8.
Cancer Gene Ther ; 28(3-4): 221-233, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32801328

RESUMEN

Long noncoding RNAs (lncRNAs) represent an emerging field of tumor biology, playing essential roles in cancer cell proliferation, invasion, and metastasis. However, the overall functional and clinical significance of most lncRNAs in pancreatic cancer is not thoroughly understood. Here, we described most of the lncRNAs with aberrant expression patterns in pancreatic cancer as detected by microarray. Quantitative real-time polymerase chain reaction further verified that the expression of LINC00671 was decreased in pancreatic cancer cell lines and patient samples. Furthermore, lower LINC00671 expression was associated with reduced tumor differentiation, aggressiveness, and poor prognosis. Functionally, LINC00671 overexpression inhibited pancreatic cancer cell proliferation, invasion, and migration in vitro, and reduced tumor growth in vivo. LINC00671 is mainly located in the cytoplasm. RNA sequencing and bioinformatics analyses indicated that LINC00671 binds to multiple miRNAs and therefore could be involved in multiple tumor-associated pathways, such as the AMPK signaling pathway and PI3K-Akt signaling pathway. Western blotting and immunohistochemistry further confirmed that LINC00671 overexpression suppressed the AKT, ERK, and epithelial-mesenchymal transition pathways. Overall, these results indicated that LINC00671 acts as a novel tumor suppressor in pancreatic cancer. Our findings may provide a new potential target for the treatment of pancreatic cancer.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Proliferación Celular/fisiología , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Largo no Codificante/genética , Análisis de Supervivencia , Transfección
9.
J Infect Dis ; 223(6): 1093-1102, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-32729925

RESUMEN

BACKGROUND: Pseudomonas aeruginosa (PA) is a pathogenic bacterium that causes severe pneumonia in critically ill and immunocompromised patients. Peptidylarginine deiminase (PAD) 2, PAD4, and caspase-1 are important enzymes in mediating host response to infection. The goal of this study was to determine the interplay between PAD2, PAD4, and caspase-1 in PA pneumonia-induced sepsis. METHODS: Pneumonia was produced in wild-type, Pad2-/-, and Pad4-/- mice by intranasal inoculation of PA (2.5 × 106 colony-forming units per mouse), and survival (n = 15/group) was monitored for 10 days. Bone marrow-derived macrophages (BMDMs) were isolated for in vitro studies. Samples were collected at specific timepoints for Western blot, bacterial load determination, and flow cytometry analysis. RESULTS: Caspase-1-dependent inflammation was diminished in PA-inoculated Pad2-/- mice, contributing to reduced macrophage death and enhanced bacterial clearance. In addition, Pad2-/- mice exhibited improved survival and attenuated acute lung injury after PA infection. In contrast, Pad4-/- mice did not display diminished caspase-1 activation, altered bacterial loads, or improved survival. CONCLUSIONS: Peptidylarginine deiminase 2 plays an essential role in the pathogenesis of pulmonary sepsis by mediating caspase-1 activation. This goes against previous findings of PAD4 in sepsis. Our study suggests that PAD2 is a potential therapeutic target of PA pneumonia-induced sepsis.


Asunto(s)
Caspasa 1 , Neumonía Bacteriana , Arginina Deiminasa Proteína-Tipo 2/metabolismo , Sepsis , Animales , Ratones , Ratones Noqueados , Neumonía Bacteriana/enzimología , Arginina Deiminasa Proteína-Tipo 4 , Pseudomonas aeruginosa , Sepsis/complicaciones , Sepsis/microbiología
10.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33055424

RESUMEN

Peptidylarginine deiminases (PADs) are a family of calcium-dependent enzymes that are involved in a variety of human disorders, including cancer and autoimmune diseases. Although targeting PAD4 has shown no benefit in sepsis, the role of PAD2 remains unknown. Here, we report that PAD2 is engaged in sepsis and sepsis-induced acute lung injury in both human patients and mice. Pad2-/- or selective inhibition of PAD2 by a small molecule inhibitor increased survival and improved overall outcomes in mouse models of sepsis. Pad2 deficiency decreased neutrophil extracellular trap (NET) formation. Importantly, Pad2 deficiency inhibited Caspase-11-dependent pyroptosis in vivo and in vitro. Suppression of PAD2 expression reduced inflammation and increased macrophage bactericidal activity. In contrast to Pad2-/-, Pad4 deficiency enhanced activation of Caspase-11-dependent pyroptosis in BM-derived macrophages and displayed no survival improvement in a mouse sepsis model. Collectively, our findings highlight the potential of PAD2 as an indicative marker and therapeutic target for sepsis.


Asunto(s)
Lesión Pulmonar Aguda/genética , Enfermedades Autoinmunes/genética , Inflamación/genética , Arginina Deiminasa Proteína-Tipo 2/genética , Sepsis/genética , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/patología , Biomarcadores/sangre , Caspasas Iniciadoras/genética , Trampas Extracelulares/genética , Regulación de la Expresión Génica/genética , Humanos , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Arginina Deiminasa Proteína-Tipo 2/antagonistas & inhibidores , Piroptosis/genética , Sepsis/sangre , Sepsis/complicaciones , Sepsis/patología
11.
Science ; 369(6510)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32943500

RESUMEN

Inflammasomes are supramolecular complexes that play key roles in immune surveillance. This is accomplished by the activation of inflammatory caspases, which leads to the proteolytic maturation of interleukin 1ß (IL-1ß) and pyroptosis. Here, we show that nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3)- and pyrin-mediated inflammasome assembly, caspase activation, and IL-1ß conversion occur at the microtubule-organizing center (MTOC). Furthermore, the dynein adapter histone deacetylase 6 (HDAC6) is indispensable for the microtubule transport and assembly of these inflammasomes both in vitro and in mice. Because HDAC6 can transport ubiquitinated pathological aggregates to the MTOC for aggresome formation and autophagosomal degradation, its role in NLRP3 and pyrin inflammasome activation also provides an inherent mechanism for the down-regulation of these inflammasomes by autophagy. This work suggests an unexpected parallel between the formation of physiological and pathological aggregates.


Asunto(s)
Histona Desacetilasa 6/metabolismo , Vigilancia Inmunológica , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pirina/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Histona Desacetilasa 6/genética , Humanos , Inflamasomas/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Centro Organizador de los Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Transporte de Proteínas
12.
J Trauma Acute Care Surg ; 89(3): 536-543, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32658440

RESUMEN

BACKGROUND: We have shown that administration of mesenchymal stem cell-derived exosomes (single dose given within 1 hour) in models of traumatic brain injury (TBI) and hemorrhagic shock is neuroprotective. The precise mechanisms responsible for the neuroprotection are not fully understood. This study was designed to investigate the transcriptomic changes in the brain that are associated with this treatment strategy. METHODS: Yorkshire swine (40-45 kg) were subjected to a severe TBI (12-mm cortical impact) and hemorrhagic shock (40% estimated total blood volume). One hour into shock, animals were randomized (n = 5/cohort) to receive either lactated Ringer's (LR; 5 mL) or exosomes suspended in LR (LR + EXO; 1 × 10 exosome particles in 5 mL LR). Animals then underwent additional shock (1 hour) followed by normal saline resuscitation. After 6 hours of observation, brain swelling (% increase compared with the uninjured side) and lesion size (mm) were assessed. Periinjured brain tissue was processed for RNA sequencing, analyzed with high through-put RNA sequencing data analysis, and results compared between control and experimental groups. RESULTS: Exosome treatment significantly increased (p < 0.005) gene expression associated with neurogenesis, neuronal development, synaptogenesis, and neuroplasticity. It also significantly reduced (p < 0.005) genes associated with stroke, neuroinflammation, neuroepithelial cell proliferation, and nonneuronal cell proliferation contributing to reactive gliosis. Exosome treatment also significantly increased (p < 0.005) the genes that are associated with stability of blood-brain barrier. CONCLUSIONS: Administration of a single dose of exosomes induces transcriptomic changes suggestive of neuroprotection. Their use as a treatment for TBI is promising and requires further investigation for human translation.


Asunto(s)
Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/terapia , Exosomas/trasplante , Células Madre Mesenquimatosas/citología , Choque Hemorrágico/terapia , Adulto , Animales , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Neuroprotección , Resucitación/métodos , Choque Hemorrágico/patología , Porcinos , Factores de Tiempo , Resultado del Tratamiento
13.
Inflammation ; 43(4): 1436-1445, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32239392

RESUMEN

Endotoxemia induced by lipopolysaccharide (LPS) is an extremely severe syndrome identified by global activation of inflammatory responses. Neutrophil extracellular traps (NETs) play an important role in the development of endotoxemia. Histone hypercitrullination catalyzed by peptidylarginine deiminases (PADs) is a key step of NET formation. We have previously demonstrated that simultaneous inhibition of PAD2 and PAD4 with pan-PAD inhibitors can decrease NETosis and improve survival in a mouse model of LPS-induced endotoxic shock. However, the effects of PAD2 specific inhibition during NETosis and endotoxic shock are poorly understood. Therefore, in the present study, we aimed to investigate the effect of the specific PAD2 or PAD4 inhibitor on LPS-induced endotoxic shock in mice. We found that PAD2 inhibition but not PAD4 inhibition improves survival. Also, the levels of proinflammatory cytokines and NETosis were significantly reduced by PAD2 inhibitor. To our knowledge, this study demonstrates for the first time that PAD2 inhibition can reduce NETosis, decrease inflammatory cytokine production, and protect against endotoxin-induced lethality. Our findings provided a novel therapeutic strategy for the treatment of endotoxic shock.


Asunto(s)
Modelos Animales de Enfermedad , Lipopolisacáridos/toxicidad , Arginina Deiminasa Proteína-Tipo 2/antagonistas & inhibidores , Arginina Deiminasa Proteína-Tipo 2/metabolismo , Choque Séptico/inducido químicamente , Choque Séptico/metabolismo , Animales , Inhibidores Enzimáticos/farmacología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Choque Séptico/mortalidad , Tasa de Supervivencia/tendencias
14.
J Trauma Acute Care Surg ; 89(2): 388-396, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32218019

RESUMEN

BACKGROUND: Early single-dose treatment with human mesenchymal stem cell-derived exosomes promotes neuroprotection and promotes blood-brain barrier integrity in models of traumatic brain injury (TBI) and hemorrhagic shock (HS) in swine. The impact of an early single dose of exosomes on late survival (7 days), however, remains unknown. We sought to evaluate the impact of early single-dose exosome treatment on neurologic outcomes, brain lesion size, inflammatory cytokines, apoptotic markers, and mediators of neural plasticity in a 7-day survival model. METHODS: Yorkshire swine were subjected to a severe TBI (8-mm cortical impact) and HS (40% estimated total blood volume). After 1 hour of shock, animals were randomized (n = 4/cohort) to receive either lactated Ringer's (5 mL) or lactated Ringer's with exosomes (1 × 10 exosome particles). After an additional hour of shock, animals were resuscitated with normal saline. Daily neurologic severity scores were compared. At 7 days following injury, lesion size, inflammatory markers, and mediators of inflammation (NF-κB), apoptosis (BAX), and neural plasticity (brain-derived neurotrophic factor) in brain tissue were compared between groups. RESULTS: Exosome-treated animals had significantly lower neurologic severity scores (first 4 days; p < 0.05) and faster neurologic recovery. At 7 days, exosome-treated animals had significantly smaller (p < 0.05) brain lesion sizes. Exosome-treated animals also had significantly lower levels of inflammatory markers (interleukin [IL]-1, IL-6, IL-8, and IL-18) and higher granulocyte-macrophage colony-stimulating factor levels compared with the control animals, indicating specific impacts on various cytokines. The BAX and NF-κB levels were significantly lower (p < 0.05) in exosome-treated animals, while brain-derived neurotrophic factor levels were significantly higher (p < 0.05) in the exosome-treated animals. CONCLUSION: In a large animal model of TBI and HS, early single-dose exosome treatment attenuates neurologic injury, decreases brain lesion size, inhibits inflammation and apoptosis, and promotes neural plasticity over a 7-day period.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/terapia , Exosomas , Neuroprotección , Choque Hemorrágico/fisiopatología , Choque Hemorrágico/terapia , Animales , Apoptosis , Barrera Hematoencefálica , Lesiones Traumáticas del Encéfalo/patología , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Hemodinámica , Inflamación/patología , Células Madre Mesenquimatosas/citología , FN-kappa B/sangre , Choque Hemorrágico/patología , Transducción de Señal , Porcinos , Resultado del Tratamiento , Proteína X Asociada a bcl-2/sangre
15.
Small ; 16(1): e1905611, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31793755

RESUMEN

Bacterial infections leading to sepsis are a major cause of deaths in the intensive care unit. Unfortunately, no effective methods are available to capture the early onset of infectious sepsis near the patient with both speed and sensitivity required for timely clinical treatment. To fill the gap, the authors develop a highly miniaturized (2.5 × 2.5 µm2 ) plasmo-photoelectronic nanostructure device that detected citrullinated histone H3 (CitH3), a biomarker released to the blood circulatory system by neutrophils. Rapidly detecting CitH3 with high sensitivity has the great potential to prevent infections from developing life-threatening septic shock. To this end, the author's device incorporates structurally engineered arrayed hemispherical gold nanoparticles that are functionalized with high-affinity antibodies. A nanoplasmonic resonance shift induces a photoconduction increase in a few-layer molybdenum disulfide (MoS2 ) channel, and it provides the sensor signal. The device achieves label-free detection of serum CitH3 with a 5-log dynamic range from 10-4 to 101 ng mL and a sample-to-answer time <20 min. Using this biosensor, the authors longitudinally measure the dynamic CitH3 profiles of individual living mice in a sepsis model at high resolution over 12 hours. The developed biosensor may be poised for future translation to personalized management of systemic bacterial infections.


Asunto(s)
Biomarcadores/metabolismo , Técnicas Biosensibles , Muerte Celular , Nanoestructuras/química , Neutrófilos/citología , Animales , Humanos , Ratones , Reproducibilidad de los Resultados
16.
Shock ; 54(4): 458-463, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31764615

RESUMEN

BACKGROUND: The peptidylarginine deiminase (PAD) family converts arginine into citrulline through protein citrullination. PAD2 and PAD4 inhibitors can improve survival in hemorrhagic shock (HS). However, the impact of isoform-specific PAD inhibition in improving survival has not been studied. In this study, we utilize selective Pad2 knockout mice to elucidate loss of function of PAD2 leads to pro-survival effect in HS. METHODS: HS: Pad2 and wild-type (WT) mice (n = 5/group) were subjected to lethal HS (55% volume hemorrhage). Survival was monitored over 7 days. Myocardial infarction (MI): Pad2 and WT mice (n = 9/group) were subjected to MI by permanent LAD ligation to examine the effect of ischemia on the heart. After 24 h cardiac function and infarct size were measured. RESULTS: HS: Pad2 mice demonstrated 100% survival compared with 0% for WT mice (P = 0.002). In a sub-lethal HS model, cardiac ß-catenin levels were higher in Pad2 compared with WT after 24 h. MI: WT mice demonstrated larger MI (75%) compared with Pad2 (60%) (P < 0.05). Pad2 had significantly higher ejection fraction and fractional shortening compared with WT (P < 0.05). CONCLUSIONS: Pad2 improves survival in lethal HS. Possible mechanisms by which loss of PAD2 function improves survival include the activation of cell survival pathways, improved tolerance of cardiac ischemia, and improved cardiac function during ischemia. PAD2 is promising as a future therapeutic target for the treatment of HS and cardiac ischemia.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Arginina Deiminasa Proteína-Tipo 2/metabolismo , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Western Blotting , Femenino , Ratones , Ratones Noqueados , Infarto del Miocardio/genética , Arginina Deiminasa Proteína-Tipo 2/genética
17.
Curr Mol Med ; 19(9): 673-682, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31339071

RESUMEN

BACKGROUND: Histone deacetylase (HDAC) 6 inhibitors have demonstrated significant protective effects in traumatic injuries. However, their roles in neuroprotection and underlying mechanisms are poorly understood. This study sought to investigate the neuroprotective effects of Tubastatin A (Tub-A), an HDAC6 inhibitor, during oxygenglucose deprivation (OGD) in HT22 hippocampal cells. METHODS: HT22 hippocampal cells were exposed to OGD. Cell viability and cytotoxicity were assessed by cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assay. Cellular apoptosis was assessed by Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Mitochondria membrane potential was detected using JC-1 dye. Expressions of acetylated α-tubulin, α-tubulin, cytochrome c, VDAC, Bax, Bcl- 2, cleaved caspase 3, phosphorylated Akt, Akt, phosphorylated GSK3ß and GSK3ß were analyzed by Western blot analysis. RESULTS: Tub-A induced acetylation of α-tubulin, demonstrating appropriate efficacy. Tub-A significantly increased cell viability and attenuated LDH release after exposure to OGD. Furthermore, Tub-A treatment blunted the increase in TUNEL-positive cells following OGD and preserved the mitochondrial membrane potential. Tub-A also attenuated the release of cytochrome c from the mitochondria into the cytoplasm and suppressed the ratio of Bax/Bcl-2 and cleaved caspase 3. This was mediated, in part, by the increased phosphorylation of Akt and GSK3ß signaling pathways. CONCLUSION: HDAC 6 inhibition, using Tub-A, protects against OGD-induced injury in HT22 cells by modulating Akt/GSK3ß signaling and inhibiting mitochondria-mediated apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucosa/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Histona Desacetilasa 6/antagonistas & inhibidores , Mitocondrias/metabolismo , Oxígeno/metabolismo , Biomarcadores , Línea Celular , Supervivencia Celular/efectos de los fármacos , Hipocampo/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Front Immunol ; 10: 2957, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998291

RESUMEN

Sepsis results in millions of deaths every year, with acute lung injury (ALI) being one of the leading causes of mortality in septic patients. As neutrophil extracellular traps (NETs) are abundant in sepsis, neutralizing components of NETs may be a useful strategy to improve outcomes of sepsis. Citrullinated histone H3 (CitH3) has been recently shown to be involved in the NET formation. In this study, we demonstrate that CitH3 damages human umbilical vein endothelial cells (HUVECs) and potentiates NET formation through a positive feedback mechanism. We developed a novel CitH3 monoclonal antibody to target peptidylarginine deiminase (PAD) 2 and PAD 4 generated CitH3. In a mouse model of lethal lipopolysaccharide (LPS) induced shock, neutralizing CitH3 with the newly developed anti-CitH3 monoclonal antibody attenuates inflammatory responses, ameliorates ALI, and improves survival. Our study suggests that effectively blocking circulating CitH3 might be a potential therapeutic method for the treatment of endotoxemia.


Asunto(s)
Histonas/inmunología , Choque Séptico/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Citrulinación , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Histonas/genética , Humanos , Masculino , Ratones , Neutrófilos/inmunología , Arginina Deiminasa Proteína-Tipo 2/inmunología , Arginina Deiminasa Proteína-Tipo 4/inmunología , Choque Séptico/tratamiento farmacológico , Choque Séptico/genética
19.
J Vis Exp ; (138)2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30199035

RESUMEN

Hemorrhage remains the leading cause of preventable deaths in trauma. Endovascular management of non-compressible torso hemorrhage has been at the forefront of trauma care in recent years. Since complete aortic occlusion presents serious concerns, the concept of partial aortic occlusion has gained a growing attention. Here, we present a large animal model of hemorrhagic shock to investigate the effects of a novel partial aortic balloon occlusion catheter and compare it with a catheter that works on the principles of complete aortic occlusion. Swine are anesthetized and instrumented in order to conduct controlled fixed-volume hemorrhage, and hemodynamic and physiological parameters are monitored. Following hemorrhage, aortic balloon occlusion catheters are inserted and inflated in the supraceliac aorta for 60 min, during which the animals receive whole-blood resuscitation as 20% of the total blood volume (TBV). Following balloon deflation, the animals are monitored in a critical care setting for 4 h, during which they receive fluid resuscitation and vasopressors as needed. The partial aortic balloon occlusion demonstrated improved distal mean arterial pressures (MAPs) during the balloon inflation, decreased markers of ischemia, and decreased fluid resuscitation and vasopressor use. As swine physiology and homeostatic responses following hemorrhage have been well-documented and are like those in humans, a swine hemorrhagic shock model can be used to test various treatment strategies. In addition to treating hemorrhage, aortic balloon occlusion catheters have become popular for their role in cardiac arrest, cardiac and vascular surgery, and other high-risk elective surgical procedures.


Asunto(s)
Aorta/fisiopatología , Choque Hemorrágico/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Porcinos
20.
Eur J Pharmacol ; 833: 432-440, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29981294

RESUMEN

Immune cell death caused by neutrophil extracellular traps (NETs), referred to as NETosis, can contribute to the pathogenesis of endotoxemia and organ damage. Although the mechanisms by which infection induces NETosis and how that leads to organ dysfunction remain largely unknown, NET formation is often found following citrullination of histone H3 (CitH3) by peptidylarginine deiminase (PAD). We hypothesized that lipopolysaccharide (LPS)-induced activation of PAD and subsequent CitH3-mediated NET formation increases endothelial permeability and pulmonary dysfunction and, therefore, that inhibition of PAD can mitigate damage and improve survival in lethal endotoxemia. Here, we showed that treatment with YW3-56, a PAD2/PAD4 inhibitor, significantly diminished PAD activation, blocked LPS-induced pulmonary vascular leakage, alleviated acute lung injury, and improved survival in a mouse model of lethal LPS-induced endotoxemia. We found CitH3 in the bloodstream 30 min after intraperitoneal injection of LPS (35 mg/kg) into mice. Additionally, CitH3 production was induced in cultured neutrophils exposed to LPS, and NETs derived from these LPS-treated neutrophils increased the permeability of endothelial cells. However, YW3-56 reduced CitH3 production and NET formation by neutrophils following LPS exposure. Moreover, treatment with YW3-56 decreased the levels of circulating CitH3 and abolished neutrophil activation and NET formation in the lungs of mice with endotoxemia. These data suggest a novel mechanism by which PAD-NET-CitH3 can play a pivotal role in pulmonary vascular dysfunction and the pathogenesis of lethal endotoxemia.


Asunto(s)
2-Naftilamina/análogos & derivados , Lesión Pulmonar Aguda/tratamiento farmacológico , Arginina/análogos & derivados , Endotoxemia/tratamiento farmacológico , Histonas/metabolismo , Desiminasas de la Arginina Proteica/antagonistas & inhibidores , 2-Naftilamina/farmacología , 2-Naftilamina/uso terapéutico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Arginina/farmacología , Arginina/uso terapéutico , Citrulinación/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotoxemia/inducido químicamente , Endotoxemia/complicaciones , Endotoxemia/mortalidad , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos/toxicidad , Pulmón/irrigación sanguínea , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Permeabilidad/efectos de los fármacos , Desiminasas de la Arginina Proteica/metabolismo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...