Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Virol ; 86(22): 11967-76, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22951825

RESUMEN

To evaluate vaccine efficacy in protecting against coxsackievirus A16 (CA16), which causes human hand, foot, and mouth disease (HFMD), we established the first neonatal mouse model. In this article, we report data concerning CA16-induced pathological changes, and we demonstrate that anti-CA16 antibody can protect mice against lethal challenge and that the neonatal mouse model could be used to evaluate vaccine efficacy. To establish a mouse model, a BJCA08/CA16 strain (at 260 50% lethal doses [LD(50)]) was isolated from a patient and used to intracerebrally (i.c.) inoculate neonatal mice. The infection resulted in wasting, hind-limb paralysis, and even death. Pathological examination and immunohistochemistry (IHC) staining indicated that BJCA08 had a strong tropism to muscle and caused severe necrosis in skeletal and cardiac muscles. We then found that BJCA08 pretreated with goat anti-G10/CA16 serum could significantly lose its lethal effect in neonatal mice. When the anti-G10 serum was intraperitoneally (i.p.) injected into the neonatal mice and, within 1 h, the same mice were intracerebrally inoculated with BJCA08, there was significant passive immunization protection. In a separate experiment, female mice were immunized with formaldehyde-inactivated G10/CA16 and BJCA08/CA16 and then allowed to mate 1 h after the first immunization. We found that there was significant protection against BJCA08 for neonatal mice born to the immunized dams. These data demonstrated that anti-CA16 antibody may block virus invasion and protect mice against lethal challenge, and that the neonatal mouse model was a viable tool for evaluating vaccine efficacy.


Asunto(s)
Enterovirus/genética , Enfermedad de Boca, Mano y Pie/virología , Vacunas/uso terapéutico , Animales , Animales Recién Nacidos , Encéfalo/patología , Chlorocebus aethiops , Medios de Cultivo , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica/métodos , Ratones , Ratones Endogámicos ICR , Modelos Genéticos , Filogenia , Reacción en Cadena de la Polimerasa , Factores de Tiempo , Vacunación , Células Vero , Carga Viral
2.
Exp Ther Med ; 21(2): 141, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33456508

RESUMEN

The progression of acute lung injury (ALI) is attributable to inflammation and oxidative stress. The cell-permeable itaconate analog 4-octyl itaconate (4-OI) provides protection against inflammatory responses and oxidative stress. However, whether 4-OI can protect against ALI remains poorly understood. The aim of this study was to explore the protective effects of 4-OI against LPS-induced ALI and the underlying mechanisms using hematoxylin and eosin (H&E) to observe lung morphology, ELISA and reverse transcription-quantitative PCR to measure the levels of IL-1ß, TNF-α and IL-6 and western blotting to examine the levels of PI3K, Akt and NF-κB. The present study demonstrates that intraperitoneal administration of 4-OI (25 mg/kg) 2 h before lipopolysaccharide (LPS; 5 mg/kg) intratracheal injection significantly alleviated the lung tissue injury induced by LPS, reducing the production of proinflammatory cytokines and reactive oxygen species (ROS) in vivo. Furthermore, 4-OI and the antioxidant N-acetyl-L-cysteine markedly suppressed PI3K and Akt phosphorylation in LPS-treated RAW264.7 macrophage cells in vitro. Further study demonstrated that a pharmacological inhibitor of the phosphoinositide 3-kinase (PI3K)-Akt pathway, LY294002, inhibited the expression of NF-κB p65 in the nuclear fraction and decreased the production of inflammatory cytokines. Collectively, the experimental results of the present study provide evidence that 4-OI significantly decreased LPS-induced lung inflammation by suppressing ROS-mediated PI3K/Akt/NF-κB signaling pathways. These results suggest that 4-OI could be a valuable therapeutic drug in the treatment of ALI.

3.
Neuroreport ; 31(8): 605-612, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32301816

RESUMEN

Subarachnoid hemorrhage (SAH) is a clinically common, acute, critical cerebrovascular disease associated with high mortality. Here, we investigated the effects of electroacupuncture on early brain injury after SAH. We successfully established a Sprague-Dawley rat model of the SAH model, and randomly divided the rats into four groups: sham-operated group, SAH group, positive control group, and electroacupuncture group. Electroacupuncture effectively decreased the number of transferase UTP nick end labeling-positive cells and extent of DNA fragmentation compared with the control, indicating a decrease in apoptosis. Moreover, electroacupuncture decreased the expression of proteins involved in the poly-ADP ribose polymerase-1/apoptosis-inducing factor (PARP-1/AIF) pathway in vivo, and the difference was statistically significant (P < 0.05). Treatment with electroacupuncture resulted in a significant improvement in neurological function. It inhibited the increase in blood-brain barrier permeability by regulating the protein expression of matrix metalloproteinase-9, occludin, and claudin-5. Additionally, electroacupuncture limited the development of cerebral edema and microglial activation in early brain injury after SAH. In conclusion, electroacupuncture can ameliorate early brain injury after SAH, and this may occur via inhibition of the PARP-1/AIF pathway.


Asunto(s)
Lesiones Encefálicas/prevención & control , Lesiones Encefálicas/fisiopatología , Electroacupuntura , Transducción de Señal , Hemorragia Subaracnoidea/complicaciones , Animales , Apoptosis , Factor Inductor de la Apoptosis/metabolismo , Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/metabolismo , Modelos Animales de Enfermedad , Microglía/fisiología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Ratas Sprague-Dawley
4.
Hum Vaccin Immunother ; 15(10): 2343-2350, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30735461

RESUMEN

Coxsackievirus A16 (CA16) has caused worldwide epidemics of hand, foot and mouth disease (HFMD), particularly in infants and pre-school children. Currently, there are no vaccines or antiviral drugs available for CA16-associated disease. In this study, a CA16-specific monoclonal antibody (MAb) NA11F12 was derived with an epidemic CA16 strain (GenBank no. JX127258). NA11F12 was found to have high cross-neutralization activity against different CA16 subgenotypes but not EV71 using RD cells. The neutralizing titers of NA11F12 ranged from 1:1024 to 1:12288 against A, B1, B2 and C subgenotypes of CA16 and was less than 8 against EV71 strain. In the neonatal mouse model, a single treatment of NA11F12 showed effective protection with a dose- and time-dependent relationship against lethal challenge by CA16 strain (GenBank no. JX481738). At day 1 post-infection, administering more than 0.1 µg/g of NA11F12 could protect 100% newborn mice from mobility and mortality challenged by CA16. With dose of 10 µg/g of NA11F12, a single administration fully protected mice against CA16-associated disease within 4 days post-infection. And there were 80% and 60% mice protected by administering NA11F12 at day 5 post-infection and day 6 post-infection when the control mice had shown clinical symptoms for 1- and 2-day, respectively. Immunohistochemical and histological analysis confirmed that NA11F12 significantly prohibited CA16 VP1 expression in various tissues and prevented CA16-induced necrosis. In conclusion, a CA16-specific MAb NA11F12 with high cross-neutralization activity was identified, which could effectively protect lethal CA16 challenge in mice. It could be a potential therapeutic MAb against CA16 in the future.


Asunto(s)
Anticuerpos Antivirales/uso terapéutico , Anticuerpos ampliamente neutralizantes/uso terapéutico , Enterovirus Humano A/inmunología , Enfermedad de Boca, Mano y Pie/prevención & control , Animales , Animales Recién Nacidos , Enfermedad de Boca, Mano y Pie/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Vacunas Virales
5.
Emerg Microbes Infect ; 7(1): 185, 2018 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-30459302

RESUMEN

As one of the key members of the coxsackievirus B group, coxsackievirus B5 (CV-B5) can cause many central nervous system diseases, such as viral encephalitis, aseptic meningitis, and acute flaccid paralysis. Notably, epidemiological data indicate that outbreaks of CV-B5-associated central nervous system (CNS) diseases have been reported worldwide throughout history. In this study, which was conducted to promote CV-B5 vaccine and anti-virus drug research, a 3-day-old BALB/c mouse model was established using a CV-B5 clinical isolate (CV-B5/JS417) as the challenge strain. Mice challenged with CV-B5/JS417 exhibited a series of neural clinical symptoms and death with necrosis of neuronal cells in the cerebral cortex and the entire spinal cord, hindlimb muscles, and cardiomyocytes. The viral load of each tissue at various post-challenge time points suggested that CV-B5 replicated in the small intestine and was subsequently transmitted to various organs via viremia; the virus potentially entered the brain through the spinal axons, causing neuronal cell necrosis. In addition, this mouse model was used to evaluate the protective effect of a CV-B5 vaccine. The results indicated that both the inactivated CV-B5 vaccine and anti-CVB5 serum significantly protected mice from a lethal infection of CV-B5/JS417 by producing neutralizing antibodies. In summary, the first CV-B5 neonatal mouse model has been established and can sustain CNS infections in a manner similar to that observed in humans. This model will be a useful tool for studies on pathogenesis, vaccines, and anti-viral drug evaluations.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Infecciones del Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/patología , Modelos Animales de Enfermedad , Animales , Animales Recién Nacidos , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/inmunología , Enterovirus Humano B , Femenino , Humanos , Intestino Delgado/virología , Ratones Endogámicos BALB C , Neuronas/patología , Neuronas/virología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Carga Viral , Viremia
6.
Zhongguo Zhen Jiu ; 37(5): 483-487, 2017 May 12.
Artículo en Zh | MEDLINE | ID: mdl-29231608

RESUMEN

OBJECTIVE: To observe the effects of electroacupuncture (EA) pretreatment on the intestinal function, inflammatory reaction and blood lactic acid in the patients of colorectal cancer surgery. METHODS: Sixty patients of colorectal cancer laparotomy at selective period, aged from 18 to 59 years old were selected. According toⅠ-Ⅲ grade of American Society of Anesthesiologists (ASA), the patients were randomized into group A (total intravenous anesthesia), group B[total intravenous anesthesia combined with EA pretreatment at Zusanli (ST 36), Shangjuxu (ST 37) and Xiajuxu (ST 39) on the right side] and group C[total intravenous anesthesia combined with EA pretreatment at Zhongwan (CV 12), Tianshu (ST 25), Zusanli (ST 36), Shangjuxu (ST 37) and Xiajuxu (ST 39) on the right side], 20 cases in each one. The total intravenous anesthesia was applied to all of the three groups. EA pretreatment was used one day before surgery and 30 min before surgery at corresponding acupoints in the group B and group C separately, 30 min each time. After surgery, the recovery time of bowel sound, the recovery time of flatus, the recovery time of defecation, the duration of diet fasting, the time of peritoneal drainage tube withdrawal and the total peritoneal drainage volume were observed in the three groups. The venous blood was collected to determine white blood cell count (WBC) and neutrophil count (NEUT) before surgery (T0), 24 h after surgery (T1) and on the 5th day after surgery (T2) separately. The arterial blood was collected to determine the level of lactic acid before surgery (TⅠ), after peritoneal douching (TⅡ) and 24 h after surgery (TⅢ)separately. RESULTS: The recovery time of bowel sound, the recovery time of flatus, the duration of diet fasting and the time of peritoneal drainage tube withdrawal in the group C were apparently earlier than those in the group A (all P<0.05). WBC and NEUT at T1 in the group C were apparently less than those in the group A (both P<0.05). The differences in lactic acid at TⅠ, TⅡ and TⅢ were not significant statistically in comparison of the three groups (all P>0.05). CONCLUSIONS: The total intravenous anesthesia combined with EA pretreatment at Zusanli (ST 36), Shangjuxu (ST 37), Xiajuxu (ST 39), Tianshu (ST 25) and Zhongwan (CV 12) reduce the recovery time of bowel sound, the recovery time of flatus, the duration of diet fasting and the time of peritoneal drainage tube withdrawal so as to improve the recovery of intestinal function, reduce WBC in 24 h after surgery and alleviate inflammatory reaction. This therapy has no influence on blood lactic acid.


Asunto(s)
Anestesia Intravenosa , Neoplasias Colorrectales/cirugía , Electroacupuntura/métodos , Puntos de Acupuntura , Adulto , Anestesia General , Neoplasias Colorrectales/fisiopatología , Humanos , Persona de Mediana Edad , Cuidados Preoperatorios , Síndrome de Respuesta Inflamatoria Sistémica/etiología
7.
J Med Virol ; 80(5): 824-32, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18360896

RESUMEN

The purpose of this study was to determine cross-protection between HEV genotypes 1 and 4, which are prevalent in China. Fecal suspensions of genotypes 1 and 4 from patients, as well as genotype 4 from swine, were inoculated intravenously into rhesus macaques. Each inoculum contained 5 x 10(4) genome equivalents of HEV. After infection, serum and fecal samples were collected serially and the levels of alanine aminotransferase (ALT) and anti-HEV IgG and IgM in sera, and HEV RNA in fecal samples, were measured. Liver biopsies were carried out. All the infected monkeys (12/12) developed anti-HEV IgG and exhibited fecal shedding of virus. IgM was detected in 11 of 12, and ALT elevation occurred about 2-6 weeks post-inoculation in 10 of 12, infected monkeys. Hepatic histopathology was consistent with acute viral hepatitis and the ORF2 antigen of HEV was detected in the granular cytoplasm of hepatocytes by immunohistochemistry. After recovery from their initial HEV infection, the monkeys were challenged with a heterologous genotype or heterologous source of HEV and monitored for hepatitis and fecal shedding. Previous infection with HEV completely or partially protected against subsequent challenge with a heterologous virus, because 7 of 11 monkeys did not develop HEV infection or shed virus in the feces, and none of them developed hepatitis or exhibited ALT elevation or liver biopsy findings of hepatitis. In conclusion, previous HEV infection may give rise to cross-genotype and cross-host-species protection.


Asunto(s)
Virus de la Hepatitis E/inmunología , Hepatitis E/inmunología , Hepatitis E/prevención & control , Alanina Transaminasa/sangre , Animales , Antígenos Virales/análisis , Reacciones Cruzadas , Heces/virología , Genotipo , Anticuerpos Antihepatitis/sangre , Virus de la Hepatitis E/genética , Hepatocitos/virología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Hígado/patología , Hígado/virología , Macaca mulatta , ARN Viral/análisis , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA