Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
1.
J Assist Reprod Genet ; 41(3): 551-561, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38200286

RESUMEN

PURPOSE: Previous studies had demonstrated that high-mobility group box 1 (HMGB1) levels were elevated in preeclampsia (PE). However, the conclusion remains controversial. This study aimed to investigate the association between blood and placenta HMGB1 levels and PE in pregnant women. METHODS: After a systematic literature search, eligible literature was screened according to inclusion and exclusion criteria. Data extraction and quality assessment were performed independently by two reviewers. The extracted data were analyzed using Review Manager 5.4 and STATA 12.0 software. Subgroup analysis and meta-regression analysis were conducted to find potential sources of heterogeneity. RESULTS: Twelve studies were included, with a total of 1145 participants. Compared with normal pregnancies, pregnant women with PE had significantly higher blood HMGB1 levels (SMD = 1.34, 95% CI: 0.72-1.95, p < 0.0001). Similarly, the expression of placental HMGB1 in PE was higher than that in normal controls by using Western blot (MD = 0.37, 95% CI: 0.27-0.47, p < 0.00001) or immunohistochemistry (OR = 6.36, 95% CI: 1.48-27.25, p = 0.01). In addition, the blood HMGB1 levels were positively correlated with the severity of PE, with higher blood HMGB1 levels in severe PE than those in mild PE (SMD = 3.35, 95% CI: 0.63-6.06, p = 0.02). The subgroup analysis indicated a close association of blood HMGB1 with PE in the Asian group, but not in the European group. CONCLUSION: Both blood and placental HMGB1 levels in patients with PE were significantly elevated, and higher blood HMGB1 levels indicated a more serious disease condition, suggesting that higher levels of HMGB1 were associated with the risk of PE.


Asunto(s)
Proteína HMGB1 , Preeclampsia , Embarazo , Humanos , Femenino , Placenta/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/análisis , Preeclampsia/genética , Preeclampsia/metabolismo , Inmunohistoquímica
2.
Blood Transfus ; 21(2): 157-167, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35302481

RESUMEN

BACKGROUND: Platelet transfusions can be associated with adverse reactions, such as febrile non-haemolytic transfusion reaction (FNHTR). It has been suggested that damage-associated molecular patterns (DAMP) and complement play a role in FNHTR. This study investigated the nature of DAMPs and complement activation products contained in platelet concentrates during storage, with a specific focus on different platelet storage solutions. MATERIALS AND METHODS: Buffy coats (BC) from healthy donors were pooled (15 BC per pool) and divided into three groups of the same volume. After addition of different storage solutions (plasma, platelet additive solutions [PAS]-C or PAS-E; n=6 for each group), BC pools were processed to platelet concentrates (PC). Leukoreduced PCs were stored on a shaking bed at 20-24°C and sampled on days 1, 2, 6 and 8 after collection for selected quality parameters: platelet activation, DAMPs (High Mobility Group Box 1 [HMGB1], nucleosomes), and complement activation products. RESULTS: During storage, equal levels of free nucleosomes and increasing concentrations of HMGB1 were present in all groups. Complement activation was observed in all PC. However, by day 8, the use of PAS had reduced C3b/c levels by approximately 90% and C4b/c levels by approximately 65%. DISCUSSION: Nucleosomes and HMGB1 were present in PCs prepared in plasma and PAS. Complement was activated during storage of platelets in plasma and in PAS. The use of PAS is associated with a lower amount of complement activation products due to the dilution of plasma by PAS . Therefore, PC in PAS have less complement activation products than platelets stored in plasma. These proinflammatory mediators in PC might induce FNHTR.


Asunto(s)
Conservación de la Sangre , Activación de Complemento , Plasma , Transfusión de Plaquetas , Soluciones , Reacción a la Transfusión , Humanos , Factores de Coagulación Sanguínea/análisis , Plaquetas , Conservación de la Sangre/efectos adversos , Conservación de la Sangre/métodos , Activación de Complemento/inmunología , Proteína HMGB1/análisis , Nucleosomas/inmunología , Activación Plaquetaria/inmunología , Transfusión de Plaquetas/efectos adversos , Transfusión de Plaquetas/métodos , Soluciones/efectos adversos , Soluciones/farmacología , Soluciones/uso terapéutico , Reacción a la Transfusión/etiología , Reacción a la Transfusión/prevención & control , Plasma/química , Plasma/inmunología , Capa Leucocitaria de la Sangre/química , Capa Leucocitaria de la Sangre/citología
3.
J Healthc Eng ; 2022: 7851436, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35392142

RESUMEN

Objective: The aim of this study is to explore the clinical application value of high-frequency ultrasound combined with detection of serum high mobility group box (HMGB-1), soluble IL-2 receptor (SIL-2R), and thyroglobulin antibody (TgAb) in diagnosing thyroid cancer. Methods: By means of retrospective study, 50 thyroid cancer patients treated in our hospital from January 2019 to January 2021 were selected as the thyroid cancer group, 50 patients with benign thyroid lesions were included in the benign lesion group, and 50 healthy individuals examined in our hospital in the same period were included in the control group. All study objects received high-frequency ultrasound examination, and at the same time, their serum HMGB-1, SIL-2R, and TgAb levels were measured. After that, the results of high-frequency ultrasound examination were analyzed, the diagnostic efficacy of different diagnosis methods was explored, and receiver operating characteristic (ROC) curves were plotted. Results: According to the results of high-frequency ultrasound examination, there were significant differences in echogenicity surrounding and inside the lesion, calcification, blood flow distribution, and blood flow parameters between the thyroid cancer group and the benign lesion group (P < 0.001); the HMGB-1, SIL-2R, and TgAb levels were statistically different among the three groups (P < 0.001), and the level values of HMGB-1, SIL-2R, and TgAb of the thyroid cancer group were, respectively, (12.26 ± 1.32) ng/ml, (108.65 ± 9.75) pmol/L, and (690.65 ± 34.47) IU/mL; the sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of high-frequency ultrasound combined with detection of serum HMGB-1, SIL-2R, and TgAb were, respectively, 98.0%, 95.0%, 90.7%, and 99.0%, and AUC (95%CI) = 0.965 (0.931-0.999). Conclusion: High-frequency ultrasound combined with detection of serum HMGB-1, SIL-2R, and TgAb has a good value in diagnosing thyroid cancer, which should be promoted in practice.


Asunto(s)
Autoanticuerpos , Proteína HMGB1 , Receptores de Interleucina-2 , Neoplasias de la Tiroides , Autoanticuerpos/análisis , Proteína HMGB1/análisis , Humanos , Receptores de Interleucina-2/análisis , Estudios Retrospectivos , Neoplasias de la Tiroides/diagnóstico por imagen , Ultrasonografía
4.
Braz. J. Pharm. Sci. (Online) ; 58: e19473, 2022. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1384023

RESUMEN

Abstract RGX-365 is the main fraction of black ginseng conmprising protopanaxatriol (PPT)-type rare ginsenosides (ginsenosides Rg4, Rg6, Rh4, Rh1, and Rg2). No studies on the antiseptic activity of RGX-365 have been reported. High mobility group box 1 (HMGB1) is recognized as a late mediator of sepsis, and the inhibition of HMGB1 release and recovery of vascular barrier integrity have emerged as attractive therapeutic strategies for the management of sepsis. In this study, we examined the effects of RGX-365 on HMGB1-mediated septic responses and survival rate in a mouse sepsis model. RGX-365 was administered to the mice after HMGB1 challenge. The antiseptic activity of RGX-365 was assessed based on the production of HMGB1, measurement of permeability, and septic mouse mortality using a cecal ligation and puncture (CLP)-induced sepsis mouse model and HMGB1-activated human umbilical vein endothelial cells (HUVECs). We found that RGX-365 significantly reduced HMGB1 release from LPS- activated HUVECs and CLP-induced release of HMGB1 in mice. RGX-365 also restored HMGB1-mediated vascular disruption and inhibited hyperpermeability in the mice. In addition, treatment with RGX-365 reduced sepsis-related mortality in vivo. Our results suggest that RGX- 365 reduces HMGB1 release and septic mortality in vivo, indicating that it is useful in the treatment of sepsis.


Asunto(s)
Proteína HMGB1/análisis , Panax/efectos adversos , Permeabilidad , Sepsis/patología , Ginsenósidos , Células Endoteliales de la Vena Umbilical Humana/clasificación , Antiinfecciosos Locales/efectos adversos
5.
Ann Hematol ; 100(9): 2293-2302, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33991204

RESUMEN

Despite the clinical value of HMGB1 in non-Hodgkin lymphoma (NHL), the impact of HMGB1 protein expression on survival of patients with mature T-cell and NK-cell lymphoma (T/NK-CL) is unknown. Here, we evaluated correlations of HMGB1 expression in tumor tissues with pathophysiological characteristics of disease and determined the prognostic value of HMGB1 expression in relapsed/refractory T/NK-CL. HMGB1 expression was detected by immunohistochemistry (IHC) in 66 cases of relapsed/refractory T/NK-CL, and specimens were classified as high or low HMGB1 expression. Univariate and multivariate Cox regression analyses identified prognostic factors associated with progression-free survival (PFS) and overall survival (OS). High HMGB1 expression was significantly correlated with increased Ki67 levels and progressive lymphoma subtypes. Univariate Cox regression analysis showed that high HMGB1 expression was associated with unfavorable PFS (P = 0.006) and poorer OS (P < 0.001). Prognostic factors identified by univariate analysis were prognostic index for peripheral T-cell lymphoma non-specified (PIT) score ≥ 2, bone marrow involvement, Ki67 ≥ 70%, and high HMGB1 expression. Multivariate Cox regression analysis revealed that high HMGB1 expression was an independent prognostic factor for poorer PFS [hazard ratio (HR) 3.593; 95% confidence interval (CI) 1.171-11.027; P = 0.025] and OS [HR 7.663; 95% CI 2.367-24.803; P = 0.001]. A proposal prognostic model combining HMGB1 and Ki67 expression showed improved prognostic capacity and may help guide treatment planning. High HMGB1 expression may be a promising prognostic predictor and a potential therapeutic target for relapsed/refractory T/NK-CL. Furthermore, to apply HMGB1 as one of the best bio-maker, an external independent control cohort is needed.


Asunto(s)
Proteína HMGB1/análisis , Linfoma Extranodal de Células NK-T/diagnóstico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclofosfamida/uso terapéutico , Doxorrubicina/uso terapéutico , Femenino , Humanos , Antígeno Ki-67/análisis , Linfoma Extranodal de Células NK-T/tratamiento farmacológico , Linfoma Extranodal de Células NK-T/patología , Linfoma Extranodal de Células NK-T/radioterapia , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/radioterapia , Prednisona/uso terapéutico , Pronóstico , Análisis de Supervivencia , Vincristina/uso terapéutico , Adulto Joven
6.
Int J Med Sci ; 18(8): 1760-1767, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746593

RESUMEN

Although high-mobility group box 1 and heat-shock protein 70 are implicated in airway diseases and suggested as relevant diagnostic biomarkers, their control concentrations in the airways have not yet been determined. This study aimed to evaluate concentration of healthy subjects for both these proteins in the upper and lower airways via meta-analysis. We searched MEDLINE, EMBASE, and Google Scholar for articles describing concentration of healthy subjects for these proteins. Data from healthy populations were combined using a random-effects model, and subgroup and sensitivity analyses were performed to determine between-study heterogeneity. We analyzed 22 studies involving 485 patients. Concentration of healthy subjects of high-mobility group box 1 and heat-shock protein 70 varied from "not detected" to 326.13 ng/mL and from 0.20 pg/mL to 9240.00 pg/mL, respectively, with the values showing significant heterogeneity. Subgroup analysis for high-mobility group box 1 revealed 13.63 ng/mL (95% CI 12.13-15.14), 100.31 ng/mL (95% CI -31.28-231.91), 9.54 ng/mL (95% CI 8.91-10.17), and 65.82 ng/mL (95% CI 55.51-76.14) for the lower airway, upper airway, pediatric populations, and adults, respectively, whereas that for heat-shock protein 70 revealed 20.58 pg/mL (95% CI 7.87-33.29) for the lower airway and 9240.00 ±11820 pg/mL for the upper airway. Although concentrations of healthy subjects of these proteins varied in the upper and lower airways, the levels of both these proteins were higher in the upper airway than in the lower airway, and these concentrations differed according to the age and sampling procedure. Our findings support the further evaluation of these proteins as biomarkers for airway-related diseases.


Asunto(s)
Proteína HMGB1/análisis , Proteínas HSP70 de Choque Térmico/análisis , Mucosa Respiratoria/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Proteína HMGB1/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Voluntarios Sanos , Humanos , Valores de Referencia
7.
FASEB J ; 35(3): e21399, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33559227

RESUMEN

The high-mobility group box-1 (HMGB1) protein is a transcription-regulating protein located in the nucleus. However, it serves as a damage-associated molecular pattern protein that activates immune cells and stimulates inflammatory cytokines to accentuate neuroinflammation after release from damaged cells. In contrast, Inter-alpha Inhibitor Proteins (IAIPs) are proteins with immunomodulatory effects including inhibition of pro-inflammatory cytokines. We have demonstrated that IAIPs exhibit neuroprotective properties in neonatal rats exposed to hypoxic-ischemic (HI) brain injury. In addition, previous studies have suggested that the light chain of IAIPs, bikunin, may exert its anti-inflammatory effects by inhibiting HMGB1 in a variety of different injury models in adult subjects. The objectives of the current study were to confirm whether HMGB1 is a target of IAIPs by investigating the potential binding characteristics of HMGB1 and IAIPs in vitro, and co-localization in vivo in cerebral cortices after exposure to HI injury. Solid-phase binding assays and surface plasmon resonance (SPR) were used to determine the physical binding characteristics between IAIPs and HMGB1. Cellular localizations of IAIPs-HMGB1 in neonatal rat cortex were visualized by double labeling with anti-IAIPs and anti-HMGB1 antibodies. Solid-phase binding and SPR demonstrated specific binding between IAIPs and HMGB1 in vitro. Cortical cytoplasmic and nuclear co-localization of IAIPs and HMGB1 were detected by immunofluorescent staining in control and rats immediately and 3 hours after HI. In conclusion, HMGB1 and IAIPs exhibit direct binding in vitro and co-localization in vivo in neonatal rats exposed to HI brain injury suggesting HMGB1 could be a target of IAIPs.


Asunto(s)
alfa-Globulinas/química , Corteza Cerebral/química , Proteína HMGB1/química , Hipoxia-Isquemia Encefálica/metabolismo , alfa-Globulinas/análisis , Animales , Animales Recién Nacidos , Femenino , Técnica del Anticuerpo Fluorescente , Proteína HMGB1/análisis , Inmunohistoquímica , Ratas , Ratas Wistar , Resonancia por Plasmón de Superficie
8.
Thorac Surg Clin ; 30(4): 395-423, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33012429

RESUMEN

Malignant pleural mesothelioma (MPM) is an asbestos-related neoplasm that can only be treated successfully when correctly diagnosed and treated early. The asbestos-exposed population is a high-risk group that could benefit from sensitive and specific blood- or tissue-based biomarkers. We review recent work with biomarker development in MPM and literature of the last 20 years on the most promising blood- and tissue-based biomarkers. Proteomic, genomic, and epigenomic platforms are covered. SMRP is the only validated blood-based biomarker with diagnostic, monitoring and prognostic value. To strengthen development and testing of MPM biomarkers, cohorts for validation must be established by enlisting worldwide collaborations.


Asunto(s)
Biomarcadores de Tumor , Mesotelioma Maligno/sangre , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/sangre , Amianto/efectos adversos , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Calbindina 2/análisis , Calbindina 2/sangre , Calbindina 2/genética , Calbindina 2/metabolismo , Proteínas de la Matriz Extracelular/análisis , Proteínas de la Matriz Extracelular/sangre , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteína HMGB1/análisis , Proteína HMGB1/sangre , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Mesotelioma Maligno/química , Mesotelioma Maligno/genética , Mesotelioma Maligno/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/análisis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Neoplasias Pleurales/sangre , Neoplasias Pleurales/química , Neoplasias Pleurales/genética , Neoplasias Pleurales/metabolismo , Pronóstico , Proteómica
9.
Life Sci ; 258: 118164, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32739467

RESUMEN

High mobility group box-1 (HMGB1) protein is a diverse, single polypeptide moiety, present in mammalian eukaryotic cells. In response to stimuli, this nuclear protein is actively secreted in to the extracellular compartment or passively released by the necrotic cells, in order to mediate inflammatory responses, by forming complexes with IL-1α, IL-1ß, LPS and other moieties, and binding to RAGE, TLR and other receptor ligands, initiating downstream, signaling processes. This molecule acts as a proinflammatory cytokine and contributes to the progression of diseases like, acute lung injury, autoimmune liver damage, graft rejection immune response and arthritis. Small concentrations of HMGB1 are released during apoptosis, which facilitates oxidative regulation on Cys106, and propagates immune inactivating tolerogenic signals in the body. The review portrays the role of HMGB1 in rheumatoid arthritis, evidently supported by pre-clinical and clinical investigations, demonstrating extensive HMGB1 expression in synovial tissue and fluid as well as serum, excessive expression of transduction receptor signaling molecules, bone remodeling and uncontrolled expression of bone destroying osteoclastogenesis, resulting in destruction of articular cartilage, bone deformation and synovial proliferation, alleviating the pathogenesis in RA disease. Moreover, the review highlights the therapeutic regime targeting HMGB1, facilitating inhibition of its actions and release into the extracellular compartment, to ameliorate the destructive events that prevail in rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/patología , Proteína HMGB1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Artritis Reumatoide/fisiopatología , Remodelación Ósea/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Cartílago Articular/patología , Cartílago Articular/fisiopatología , Proteína HMGB1/análisis , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Terapia Molecular Dirigida , Osteogénesis/efectos de los fármacos
10.
PLoS One ; 15(6): e0235220, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32584885

RESUMEN

BACKGROUND: Approximately 30,000 patients with blunt cardiac trauma are recorded each year in the United States. Blunt cardiac injuries after trauma are associated with a longer hospital stay and a poor overall outcome. Organ damage after trauma is linked to increased systemic release of pro-inflammatory cytokines and damage-associated molecular patterns. However, the interplay between polytrauma and local cardiac injury is unclear. Additionally, the impact of surgical intervention on this process is currently unknown. This study aimed to determine local cardiac immunological and structural alterations after multiple trauma. Furthermore, the impact of the chosen fracture stabilization strategy (reamed versus non-reamed femoral nailing) on cardiac alterations was studied. EXPERIMENTAL APPROACH: 15 male pigs were either exposed to multiple trauma (blunt chest trauma, laparotomy, liver laceration, femur fracture and haemorrhagic shock) or sham conditions. Blood samples as well as cardiac tissue were analysed 4 h and 6 h after trauma. Additionally, murine HL-1 cells were exposed to a defined polytrauma-cocktail, mimicking the pro-inflammatory conditions after multiple trauma in vitro. RESULTS: After multiple trauma, cardiac structural changes were observed in the left ventricle. More specifically, alterations in the alpha-actinin and desmin protein expression were found. Cardiac structural alterations were accompanied by enhanced local nitrosative stress, increased local inflammation and elevated systemic levels of the high-mobility group box 1 protein. Furthermore, cardiac alterations were observed predominantly in pigs that were treated by non-reamed intramedullary reaming. The polytrauma-cocktail impaired the viability of HL-1 cells in vitro, which was accompanied by a release of troponin I and HFABP. DISCUSSION: Multiple trauma induced cardiac structural alterations in vivo, which might contribute to the development of early myocardial damage (EMD). This study also revealed that reamed femoral nailing (reamed) is associated with more prominent immunological cardiac alterations compared to nailing without reaming (non-reamed). This suggests that the choice of the initial fracture treatment strategy might be crucial for the overall outcome as well as for any post-traumatic cardiac consequences.


Asunto(s)
Clavos Ortopédicos/efectos adversos , Fracturas del Fémur/cirugía , Traumatismo Múltiple/patología , Miocardio/patología , Actinina/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Supervivencia Celular , Conexina 43/metabolismo , Citocinas/análisis , Citocinas/metabolismo , Desmina/metabolismo , Fracturas del Fémur/patología , Proteína HMGB1/análisis , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Inflamación , Masculino , Ratones , Traumatismo Múltiple/metabolismo , Traumatismo Múltiple/veterinaria , Miocardio/metabolismo , Estrés Nitrosativo , Porcinos , Troponina I/análisis
11.
Biochem Biophys Res Commun ; 530(2): 455-461, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32553625

RESUMEN

Osteoclasts are multinucleated bone-resorbing cells derived from monocyte/macrophage progenitor cells. Excessive formation and resorbing activities of osteoclasts are involved in the bone-destructive pathologies of rheumatoid arthritis and osteoporosis. Recently, it has been found that nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor for anti-oxidative stress genes, functions in osteoclastogenesis. Dimethyl fumarate (DMF) is a potent activator of Nrf2 and has been shown to inhibit osteoclastogenesis. Here, we investigated the mechanisms of this inhibition by examining the activation of several signalling pathways during the differentiation of bone marrow-derived macrophages into osteoclasts. DMF inhibited the differentiation of osteoclasts in a dose-dependent manner and suppressed the bone-resorbing activity of osteoclasts. DMF treatment decreased the expression of nuclear factor of activated T-cells cytoplasmic-1, and significantly decreased phosphorylation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase in osteoclasts. We also found that DMF inhibited the extracellular release of high mobility group box 1, associated with an up-regulation of heme oxygenase-1, likely mediated through Nrf2 activation. Our results indicate that DMF inhibits osteoclast differentiation through multiple pathways.


Asunto(s)
Dimetilfumarato/farmacología , Proteína HMGB1/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Osteogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Animales , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína HMGB1/análisis , Masculino , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Biochem Biophys Res Commun ; 527(3): 839-844, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32430176

RESUMEN

Apoptosis is the prototype for a regulated form of cell death, but recent studies have revealed other types of regulated forms of cell death, including necroptosis and ferroptosis. The molecular mechanisms underlying the execution of these processes have been intensively investigated, yet the hallmarks of their morphology are not fully understood. Here, we report that electron lucent cytoplasm was a common feature of both necroptosis and ferroptosis, which was consistent with cytoplasmic vacuolization due to a defect in the cytoplasmic membrane integrity. Notably, the perinuclear space was dilated in necroptosis, but such dilation did not occur in ferroptosis. Cells undergoing ferroptosis, but not necroptosis, exhibited an electron lucent nucleus. We previously reported that one of the nuclear danger-associated molecular patterns (DAMPs), high mobility group box (HMGB)1, is rapidly released from the nucleus to the extracellular spaces of cells undergoing necroptosis through the ruptured nuclear and cytoplasmic membrane. Via time-lapse imaging of cells stably expressing HMGB1 fused to a fluorescence protein, we found that HMGB1 was also released from the nucleus to the cytosol, and then eventually released into the extracellular spaces in cells undergoing ferroptosis. Thus, nuclear membrane damage was induced prior to cytoplasmic membrane rupture in ferroptosis. Thus, dilation of the perinuclear space and an electron lucent nucleus may be the hallmarks of necroptosis and ferroptosis, respectively.


Asunto(s)
Ferroptosis , Necroptosis , Línea Celular Tumoral , Membrana Celular/ultraestructura , Núcleo Celular/ultraestructura , Citoplasma/ultraestructura , Proteína HMGB1/análisis , Humanos , Microscopía Electrónica de Transmisión/métodos
13.
Medicine (Baltimore) ; 98(50): e18322, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31852121

RESUMEN

BACKGROUND: Drug induced liver injury (DILI) is an increasing cause of acute liver injury especially with increasing need for pharmacotherapy of widening comorbidities amongst our ever-aging population. Uncertainty however remains regarding both acceptable and widely agreeable diagnostic algorithms as well a clear understanding of mechanistic insights that most accurately underpins it. In this review, we have explored the potential role of emerging novel markers of DILI and how they could possibly be integrated into clinical care of patients. METHODS: We explored PUBMED and all other relevant databases for scientific studies that explored potential utility of novel biomarkers of DILI, and subsequently carried out a narrative synthesis of this data. As this is a narrative review with no recourse to patient identifiable information, no ethics committee's approval was sought or required. RESULTS: Novel biomarkers such as microRNA-122 (miR-122) profiles, high mobility group box-1 (HMGB1), glutamate dehydrogenase (GLDH), and cytokeratin-18 (K-18), amongst others do have the potential for reducing diagnostic uncertainties associated with DILI. CONCLUSION: With the increasing validation of some of the novel liver biomarkers such as K-18, mir-122, HMGB-1, and GLDH, there is the potential for improvement in the diagnostic uncertainty commonly associated with cases of DILI.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Glutamato Deshidrogenasa/análisis , Proteína HMGB1/análisis , Queratina-18/análisis , MicroARNs/análisis , Medición de Riesgo/métodos , Biomarcadores/análisis , Humanos
14.
Eur Rev Med Pharmacol Sci ; 23(24): 10950-10956, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31858563

RESUMEN

OBJECTIVE: To investigate the influences of micro ribonucleic acid (miR)-205 on renal injury in sepsis rats through the high-mobility group box 1 (HMGB1)-phosphatase and tensin homolog deleted on chromosome ten (PTEN) signaling pathway. MATERIALS AND METHODS: A rat model of sepsis-induced renal injury was established by cecal ligation and perforation. The rats were randomly divided into 3 groups, namely the Sham group, the Model group, and the miR-205 group. Hematoxylin and eosin (HE) staining was applied to examine the pathological renal morphology. The enzyme-linked immunosorbent assay (ELISA) was adopted to measure the serum levels of Caspase-3 and Bcl-2-associated X protein (Bax) in rats. Cell apoptosis rate in the renal tissues was detected via terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Finally, the protein levels of phosphorylate-HMGB1 (p-HMGB1) and p-PTEN in the renal tissues were determined using the Western blotting (WB) assay. RESULTS: Compared with those in the Sham group, the pathological morphology of the renal tissues was poor in Model group. The serum levels of Caspase-3 and Bax, the apoptosis rate, and the protein levels of p-HMGB1 and p-PTEN were remarkably enhanced in the Model group compared to the Sham group. In comparison with those in Model group, the pathological changes in renal morphology, apoptosis-related indexes, and protein levels of p-HMGB1 and p-PTEN were alleviated in the miR-205 group. CONCLUSIONS: MiR-205 agonist can improve the pathological morphology in the sepsis rats with renal injury, improve renal cell apoptosis, and inhibit the protein levels of HMGB1 and PTEN in renal tissues. MiR-205 alleviates sepsis-induced renal injury through the HMGB1-PTEN signaling pathway.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteína HMGB1/metabolismo , MicroARNs/metabolismo , Fosfohidrolasa PTEN/metabolismo , Sepsis/metabolismo , Transducción de Señal , Lesión Renal Aguda/patología , Animales , Apoptosis , Proliferación Celular , Modelos Animales de Enfermedad , Proteína HMGB1/análisis , Masculino , Fosfohidrolasa PTEN/análisis , Fosforilación , Ratas , Ratas Sprague-Dawley , Sepsis/patología
15.
Methods Enzymol ; 629: 177-193, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31727240

RESUMEN

The exodus of the alarmin high mobility group box 1 (HMGB1) from the nucleus constitutes a crucial cellular danger signal and manifests as a sequential process in which HMGB1 first exits the nucleus into the cytoplasm and then is secreted or passively released through the permeabilized plasma membrane. Extracellular HMGB1 can interact with pattern recognition receptors to stimulate innate immune responses. Here, we describe a discovery pipeline for the identification of pharmacological agents endowed with HMGB1 releasing properties. The "retention using selective hooks" (RUSH) system in which a streptavidin-NLS3 fusion protein serves as a nuclear hook to sequester streptavidin-binding peptide (SBP) fused with HMGB1 and green fluorescent protein (GFP) allowed for synchronizing HMGB1 increase. Thus, exclusively in the presence of biotin, which liberates HMGB1-SBP-GFP from its nuclear hook, immunogenic cell death (ICD) inducers such as anthracyclines are able to cause the nucleo-cytoplasmic translocation of HMGB1-SBP-GFP. This system facilitates the identification of HMGB1 releasing agents in medium- to high-throughput screening assays.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas/métodos , Proteína HMGB1/análisis , Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Membrana Celular/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Descubrimiento de Drogas/instrumentación , Proteínas Fluorescentes Verdes/genética , Proteína HMGB1/inmunología , Proteína HMGB1/metabolismo , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Inmunidad Innata/efectos de los fármacos , Muerte Celular Inmunogénica/efectos de los fármacos , Sondas Moleculares/genética , Neoplasias/inmunología , Neoplasias/patología , Señales de Localización Nuclear/genética , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Estreptavidina/genética
16.
Methods Enzymol ; 629: 195-217, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31727241

RESUMEN

HMGB1 is the most abundant non-histone nuclear protein. It regulates transcriptional access to open areas of chromatin and limits release of DNA with apoptotic death, serving to both inhibit apoptosis and promote DNA repair. When HMGB1 is translocated to the cytosol with many types of cellular stress, it is a powerful inducer of autophagy. It can also be released by activated immune cells and damaged or dying cells into the extracellular space, where it acts as a damage associated molecular pattern (DAMP) molecule, contributing to the pathogenesis and progression of cancer. Here, the most common methodologies to not only measure HMGB1 but also to effectively determine its subcellular localization, which dictates many of HMGB1's different functions, are reviewed.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteína HMGB1/análisis , Neoplasias/inmunología , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Autofagia/inmunología , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Carcinogénesis/patología , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Citosol/inmunología , Citosol/metabolismo , Progresión de la Enfermedad , Espacio Extracelular/inmunología , Espacio Extracelular/metabolismo , Proteína HMGB1/inmunología , Proteína HMGB1/metabolismo , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Muerte Celular Inmunogénica/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Microambiente Tumoral/inmunología
17.
AIDS ; 33(11): 1683-1692, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31149942

RESUMEN

OBJECTIVE: To use plasma neuron-derived exosomes (NDEs) to detect proteins that diagnose HIV-associated neurocognitive disorders (HAND). To compare NDE cargo from HAND with Alzheimer's disease. DESIGN: Eighty plasma samples were assayed including men (n = 29) and women (n = 51) with and without HAND. METHODS: Plasma NDEs were isolated by immunoadsorption with neuron specific L1 cell adhesion molecule antibody. NDE proteins were quantified by ELISA and proximity extension assays for 184 targets. RESULTS: Neuronal enrichment of NDE was confirmed with elevated synaptophysin and normalized to the exosomal marker, apoptosis-linked gene-2-interacting protein X (ALIX). NDE from men and women had significant divergent results. High mobility group box 1 and neurofilament light were significantly increased in NDE from cognitively impaired men and were unchanged in women. NDE from HIV+ men had decreased p-T181-tau, a marker increased in Alzheimer's disease, compared with no difference in women. NDE amyloid beta was not increased in cognitive impairment. Proximity extension assays analysis showed 25 proteins were differentially expressed in HIV infection alone. Seven proteins identified asymptomatic and mild cognitive impairment in HIV+ women. NDE from women had significantly decreased cathepsin S, total tau, neuronal cell adhesion molecule and contactin 5 in mild impairment. Twelve proteins were increased in NDE from cognitively impaired men, including carboxypeptidase M, cadherin 3, colony stimulating factor 2 receptor alpha subunit and mesencephalic astrocyte-derived neurotropic factor. CONCLUSION: NDE proteins differ in HIV infection alone and cognitive impairment between men and women suggesting mechanistic sex differences associated with HAND. Several NDE targets are different from that reported for Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/análisis , Disfunción Cognitiva/diagnóstico , Exosomas/química , Infecciones por VIH/complicaciones , Proteína HMGB1/análisis , Proteínas de Neurofilamentos/análisis , Complejo SIDA Demencia/diagnóstico , Complejo SIDA Demencia/patología , Adulto , Biomarcadores/sangre , Análisis Químico de la Sangre , Disfunción Cognitiva/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad
18.
Medicina (Kaunas) ; 55(5)2019 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-31137593

RESUMEN

Acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) is an acute respiratory worsening of unidentifiable cause that sometimes develops during the clinical course of IPF. Although the incidence of AE-IPF is not high, prognosis is poor. The pathogenesis of AE-IPF is not well understood; however, evidence suggests that coagulation abnormalities and inflammation are involved. Thrombomodulin is a transmembranous glycoprotein found on the cell surface of vascular endothelial cells. Thrombomodulin combines with thrombin, regulates coagulation/fibrinolysis balance, and has a pivotal role in suppressing excess inflammation through its inhibition of high-mobility group box 1 protein and the complement system. Thus, thrombomodulin might be effective in the treatment of AE-IPF, and we and other groups found that recombinant human soluble thrombomodulin improved survival in patients with AE-IPF. This review summarizes the existing evidence and considers the therapeutic role of thrombomodulin in AE-IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Trombomodulina/uso terapéutico , Biomarcadores/análisis , Biomarcadores/sangre , Progresión de la Enfermedad , Proteína HMGB1/análisis , Proteína HMGB1/sangre , Humanos , Fibrosis Pulmonar Idiopática/fisiopatología , Pronóstico , Resultado del Tratamiento
19.
Anal Chem ; 91(9): 6035-6042, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30990031

RESUMEN

A new proteomic strategy combining functionalized magnetic nanoparticle affinity probes with mass spectrometry was developed to capture and identify proteins specifically responding to 1,2-d(GpG) intrastrand cisplatin-cross-linked DNA, the major DNA lesion caused by cisplatin and thought to induce apoptosis. A 16-mer oligodeoxynucleotide (ODN) duplex and its cisplatin-cross-linked adduct were immobilized on magnetic nanoparticles via click reaction, respectively, to fabricate negative and positive affinity probes which were very stable in cellular protein extracts due to the excellent bio-orthogonality of click chemistry and the inertness of covalent triazole linker. Quantitative mass spectrometry results unambiguously revealed the predominant binding of HMGB1 and HMGB2, the well-established specific binders of 1,2-cisplatin-cross-linked DNA, to the cisplatin-cross-linked ODN, thus validating the accuracy and reliability of our strategy. Furthermore, 5 RNA or single-stranded DNA binding proteins, namely, hnRNP A/B, RRP44, RL30, RL13, and NCL, were demonstrated to recognize specifically the cisplatinated ODN, indicating the significantly unwound ODN duplex by cisplatin cross-linking. In contrast, the binding of a transcription factor TFIIFa to DNA was retarded due to cisplatin damage, implying that the cisplatin lesion stalls DNA transcription. These findings promote understanding in the cellular responses to cisplatin-damaged DNA and inspire further precise elucidation of the action mechanism of cisplatin.


Asunto(s)
Cisplatino/farmacología , ADN/efectos de los fármacos , Proteína HMGB1/análisis , Proteína HMGB2/análisis , Proteómica , Daño del ADN , Humanos , Células MCF-7 , Nanopartículas de Magnetita/química , Espectrometría de Masas , Estructura Molecular , Células Tumorales Cultivadas
20.
PLoS One ; 14(4): e0211756, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30964875

RESUMEN

Rapid Diagnostic Tests (RDTs) for malaria are restricted to a few biomarkers and antibody-mediated detection. However, the expression of commonly used biomarkers varies geographically and the sensibility of immunodetection can be affected by batch-to-batch differences or limited thermal stability. In this study we aimed to overcome these limitations by identifying a potential biomarker and by developing molecular sensors based on aptamer technology. Using gene expression databases, ribosome profiling analysis, and structural modeling, we find that the High Mobility Group Box 1 protein (HMGB1) of Plasmodium falciparum is highly expressed, structurally stable, and present along all blood-stages of P. falciparum infection. To develop biosensors, we used in vitro evolution techniques to produce DNA aptamers for the recombinantly expressed HMG-box, the conserved domain of HMGB1. An evolutionary approach for evaluating the dynamics of aptamer populations suggested three predominant aptamer motifs. Representatives of the aptamer families were tested for binding parameters to the HMG-box domain using microscale thermophoresis and rapid kinetics. Dissociation constants of the aptamers varied over two orders of magnitude between nano- and micromolar ranges while the aptamer-HMG-box interaction occurred in a few seconds. The specificity of aptamer binding to the HMG-box of P. falciparum compared to its human homolog depended on pH conditions. Altogether, our study proposes HMGB1 as a candidate biomarker and a set of sensing aptamers that can be further developed into rapid diagnostic tests for P. falciparum detection.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Proteína HMGB1/análisis , Plasmodium falciparum/aislamiento & purificación , Proteínas Protozoarias/análisis , Secuencia de Aminoácidos , Secuencia de Bases , Humanos , Malaria Falciparum/diagnóstico , Malaria Falciparum/parasitología , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA