Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Infect Public Health ; 17(1): 163-171, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38039859

RESUMEN

BACKGROUND: Several genital pathogens affect fertility. The study estimated the seroprevalence of Treponema pallidum, Ureaplasma urealyticum, and Mycoplasma hominis and identify specific factors associated with exposure to at least one of these pathogens in patients seeking fertility treatment in the Emirate of Abu Dhabi, United Arab Emirates. METHODS: A seroepidemiological survey was conducted in a major fertility clinic in the Emirate of Abu Dhabi. Serum samples were screened for eight immunoglobulins (IgG, IgM, and IgA) against T. pallidum, U. urealyticum, and M. hominis using enzyme-linked immunoassays. Factors associated with seropositivity to at least one of the pathogens were investigated. RESULTS: The study surveyed 308 patients seeking fertility treatment (mean age: 36.1 ± 6.8 years). Most patients were female (88.0%), 24.9% had at least one chronic comorbidity, 19.3% had a previous genital infection, and 68.1% had been diagnosed with infertility for ≥ 6 months. Ig seroprevalence of T. pallidum (IgG: 3.0%, IgM: 3.2%), U. urealyticum (IgG: 2.6%, IgM: 2.0%), and M. hominis (IgG: 33.9%) was 6.4%, 4.6%, and 49.0%, respectively. Nearly one quarter (23.0%) and one decile (9.2%) of the patients exhibited evidence of ongoing infection (IgM seropositivity) or recent infection (IgA seropositivity) with M. hominis, respectively. Overall, 53.0% of the patients were seropositive for at least one of the screened immunoglobulins. Patients with an education level of secondary schooling or below (66.2%) or those who were unemployed (61.1%) had a higher seroprevalence of IgG antibodies compared with patients with college or higher-level education (48.4%) or those who were employed (48.1%) (p < 0.05). CONCLUSION: Exposure to T. pallidum or U. urealyticum was relatively low, whereas that to M. hominis was common in the surveyed patients. Enhanced awareness and screening programmes for genital pathogens are crucial to prevent and control the transmission of infections and reduce the growing burden of infertility.


Asunto(s)
Infertilidad , Ureaplasma urealyticum , Humanos , Femenino , Adulto , Masculino , Mycoplasma hominis , Emiratos Árabes Unidos/epidemiología , Treponema pallidum , Estudios Seroepidemiológicos , Infertilidad/epidemiología , Inmunoglobulina G , Inmunoglobulina A , Inmunoglobulina M
2.
Sex Transm Dis ; 50(9): 583-590, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37277894

RESUMEN

PURPOSE: This study was designed to investigate the seroepidemiology of and identify factors associated with exposure to Chlamydia trachomatis ( C. trachomatis ) in fertility treatment-seeking patients in Abu Dhabi Emirate, United Arab Emirates. METHODS: A total of 308 fertility treatment-seeking patients were surveyed. Seroprevalence of past (IgG positive), current/acute (IgM positive), and active infection (IgA positive) with C. trachomatis was quantified. Factors associated with exposure to C. trachomatis were identified. RESULTS: Overall, 19.0%, 5.2%, and 1.6% found to have past, acute/recent, and ongoing active infection with C. trachomatis , respectively. Overall, 22.0% of the patients were seropositive to any of the 3 to C. trachomatis antibodies. Male compared with female patients (45.7% vs. 18.9%, P < 0.001) and current/ex-smokers compared with nonsmokers (44.4% vs. 17.8%) had higher seropositivity. Patients with a history of pregnancy loss had higher seropositivity compared with other patients (27.0% vs. 16.8%), particularly recurrent pregnancy losses (33.3%). Current smoking (adjusted odds ratio [aOR], 3.8; 95% confidence interval, 1.32-11.04) and history of pregnancy loss (adjusted odds ratio [aOR], 3.0; 95% confidence interval, 1.5-5.8) were significantly associated with higher odds of exposure to C. trachomatis . CONCLUSIONS: The observed high seroprevalence of C. trachomatis , particularly in patients with a history of pregnancy loss, possibly indicates the contribution of C. trachomatis to the growing burden of infertility in the United Arab Emirates.


Asunto(s)
Infecciones por Chlamydia , Infertilidad , Embarazo , Humanos , Masculino , Femenino , Chlamydia trachomatis , Emiratos Árabes Unidos/epidemiología , Estudios Seroepidemiológicos , Infertilidad/epidemiología , Proyectos de Investigación , Infecciones por Chlamydia/epidemiología , Infecciones por Chlamydia/complicaciones
3.
Front Public Health ; 10: 991040, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36420003

RESUMEN

Background: Herpes simplex virus type 2 (HSV-2) is a common genitally-transmitted viral infection affecting more than 400 million individuals globally. In the United Arab Emirates (UAE), in specific at-risk population groups, the burden of HSV-2 has not been reported. This study investigated the prevalence of HSV-2 IgG antibodies in patients seeking fertility treatment and characterized patients with seropositivity to HSV-2 IgG antibodies. Methodology: A cross-sectional sample of patients seeking fertility treatment in a major fertility clinic in Abu Dhabi, UAE was surveyed from April to May 2021. Patients were consecutively invited to complete self-administered questionnaires and provide blood for HSV-2 testing. Information on sociodemographics, medical history, and infertility was collected. Serum specimens were screened using an enzyme-linked immunosorbent assay for HSV-2 IgG antibodies detection. Results: Two hundred and ninety-nine patients were surveyed and provided blood samples. The mean age of the patients was 35.9 ± 6.8 [mean ± standard deviation (SD)] years with 89.3% being women. Sixty-six percent were overweight or obese, 25.0% had at least one chronic comorbidity, and 19.6% reported ever-had genital infection. More than two-thirds (68.3%) of the patients were infertile for ≥ 6 months. Of the 42 infertile males, 69.0% had an abnormal semen analysis. HSV-2 IgG antibodies was detected in 12.4% of patients. The HSV-2 IgG seropositive patients had a higher mean age (39.5 vs. 35.4 years; p < 0.001) compared to seronegative patients. HSV-2 IgG antibodies seropositivity was more common in males (15.6%) than females (12.0%), in patients with secondary (14.1%) vs. primary (9.2%) infertility, or in males with abnormal (10.3%) vs. normal (7.7%) semen. Conclusion: Exposure to HSV-2 at any time in patients seeking fertility treatment in the UAE was found to be slightly common in more than one out of 10 patients. Tailored health campaigns on HSV-2 prevention are warranted.


Asunto(s)
Herpes Genital , Infertilidad , Masculino , Humanos , Femenino , Herpesvirus Humano 2 , Estudios Seroepidemiológicos , Estudios Transversales , Herpes Genital/epidemiología , Herpes Genital/diagnóstico , Emiratos Árabes Unidos/epidemiología , Anticuerpos Antivirales , Inmunoglobulina G
4.
Viruses ; 13(8)2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34452495

RESUMEN

Influenza A virus (IAV) and respiratory syncytial virus (RSV) are leading causes of childhood infections. RSV and influenza are competitive in vitro. In this study, the in vivo effects of RSV and IAV co-infection were investigated. Mice were intranasally inoculated with RSV, with IAV, or with both viruses (RSV+IAV and IAV+RSV) administered sequentially, 24 h apart. On days 3 and 7 post-infection, lung tissues were processed for viral loads and immune cell populations. Lung functions were also evaluated. Mortality was observed only in the IAV+RSV group (50% of mice did not survive beyond 7 days). On day 3, the viral loads in single-infected and co-infected mice were not significantly different. However, on day 7, the IAV titer was much higher in the IAV+RSV group, and the RSV viral load was reduced. CD4 T cells were reduced in all groups on day 7 except in single-infected mice. CD8 T cells were higher in all experimental groups except the RSV-alone group. Increased airway resistance and reduced thoracic compliance were demonstrated in both co-infected groups. This model indicates that, among all the infection types we studied, infection with IAV followed by RSV is associated with the highest IAV viral loads and the most morbidity and mortality.


Asunto(s)
Coinfección/virología , Virus de la Influenza A/fisiología , Gripe Humana/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/mortalidad , Gripe Humana/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/mortalidad , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitial Respiratorio Humano/genética , Índice de Severidad de la Enfermedad , Carga Viral
5.
Eur J Immunol ; 50(7): 1000-1018, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32125695

RESUMEN

Dengue infection causes dengue fever (DF) and dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). CD4+ Foxp3+ Tregs are expanded in patients during dengue infection, and appear to be associated with clinical severity. However, molecular pathways involved in Treg proliferation and the reason for their insufficient control of severe diseases are poorly understood. Here, dengue infection induced the proliferation of functional CD4+ Foxp3+ Tregs via TLR2/MyD88 pathway. Surface TLR2 on Tregs was responsible for their proliferation, and dengue-expanded Tregs subverted in vivo differentiation of effector CD8+ T cells. An additional interesting finding was that dengue-infected hosts displayed changed levels of susceptibility to other diseases in TLR2-dependent manner. This change included enhanced susceptibility to tumors and bacterial infection, but highly enhanced resistance to viral infection. Further, the transfer of dengue-proliferated Tregs protected the recipients from dengue-induced DHF/DSS and LPS-induced sepsis. In contrast, dengue-infected hosts were more susceptible to sepsis, an effect attributable to early TLR2-dependent production of proinflammatory cytokines. These facts may explain the reason why in some patients, dengue-proliferated Tregs is insufficient to control DF and DHF/DSS. Also, our observations lead to new insights into Treg responses activated by dengue infection in a TLR2-dependent manner, which could differentially act on subsequent exposure to other disease-producing situations.


Asunto(s)
Virus del Dengue/inmunología , Dengue/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 2/inmunología , Enfermedad Aguda , Animales , Línea Celular Tumoral , Dengue/patología , Ratones , Linfocitos T Reguladores/patología
6.
J Chemother ; 32(2): 88-97, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31884896

RESUMEN

Idelalisib, an inhibitor of the phosphatidylinositol-3-kinase p110δ subunit (PI3Kδ), is approved for treating lymphoid malignancy. The drug is associated with hematopoietic and pulmonary toxicities, which limit its clinical use. However, the toxicity mechanisms are not completely elucidated. In this study, mice were intraperitoneally injected with idelalisib (40 or 80 µg/g) or dimethyl sulfoxide for five days every week for up to four weeks to evaluate the changes in the thymus, spleen, and pulmonary functions. Idelalisib treatment induced thymic involution, decreased CD4+/CD8+ T-cell population, and increased CD4-/CD8- T-cell population. In the spleen, idelalisib dose dependently decreased the lymphocyte viability and cell count. Idelalisib-treated mice exhibited enhanced cleaved caspase-3 expression in the thymus, spleen, and lung tissues. Idelalisib augmented thoracic and airway resistance and decreased thoracic compliance. Thus, PI3Kδ has physiological roles in T-cell development and airway function. Monitoring drug toxicity is important for developing follow-up compounds that target PI3Kδ signalling.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Tejido Linfoide/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Intraperitoneales , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Purinas , Quinazolinonas , Transducción de Señal/efectos de los fármacos , Bazo/efectos de los fármacos , Timo/efectos de los fármacos
7.
Int J Infect Dis ; 89: 96-101, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31493522

RESUMEN

OBJECTIVE: As the current recommendation of administering Tdap (tetanus-diphtheria-acellular pertussis) to all pregnant women has not been widely implemented in the United Arab Emirates (UAE), we aimed to ascertain the prevalence of pertussis seronegativity during pregnancy. METHODS: IgG antibodies against Bordetella pertussis toxin (PT) were measured in 213 women attending the antenatal clinic at Oasis hospital, Al Ain, UAE. Results were compared by maternal age, nationality and gestational age with the Kruskal-Wallis test for IgG-PT levels and the Chi-squared test for serology status. RESULTS: The mean age±SD of the participants was 30.4±5.6 years, mean gestational age±SD of 25.5±3.3 weeks. Serum concentration of IgG-PT <10IU/ml were found in 160 out of 213 women (75%; 95% confidence interval 69%, 81%). There was no significant difference in the geometric mean of serum IgG-PT concentration across maternal age (P=0.80) or nationality (P=0.90). There were no differences in the prevalence of seronegativity with maternal age (P=0.65) or nationality (P=0.90). CONCLUSION: With a high prevalence of pertussis seronegativity in pregnant women, there is a potential benefit of introducing pertussis vaccination during pregnancy into our national immunization program.


Asunto(s)
Complicaciones del Embarazo/sangre , Tos Ferina/sangre , Adulto , Anticuerpos Antibacterianos/sangre , Bordetella pertussis/inmunología , Femenino , Edad Gestacional , Humanos , Programas de Inmunización , Inmunoglobulina G/sangre , Embarazo , Complicaciones del Embarazo/diagnóstico , Complicaciones del Embarazo/microbiología , Complicaciones del Embarazo/prevención & control , Emiratos Árabes Unidos , Vacunación/métodos , Tos Ferina/diagnóstico , Tos Ferina/microbiología , Tos Ferina/prevención & control , Adulto Joven
8.
Front Immunol ; 10: 1038, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156627

RESUMEN

Type I diabetes (T1D) is a T cell-driven autoimmune disease that results in the killing of pancreatic ß-cells and, consequently, loss of insulin production. Using the multiple low-dose streptozotocin (MLD-STZ) model of experimental autoimmune diabetes, we previously reported that pretreatment with a specific acetylcholinesterase inhibitor (AChEI), paraoxon, prevented the development of hyperglycemia in C57BL/6 mice. This correlated with an inhibition of T cell infiltration into the pancreatic islets and a reduction in pro-inflammatory cytokines. The cholinergic anti-inflammatory pathway utilizes nicotinic and muscarinic acetylcholine receptors (nAChRs and mAChRs, respectively) expressed on a variety of cell types. In this study, we carried out a comparative analysis of the effect of specific antagonists of nAChRs or mAChRs on the development of autoimmune diabetes. Co-administration of mecamylamine, a non-selective antagonist of nAChRs maintained the protective effect of AChEI on the development of hyperglycemia. In contrast, co-administration of atropine, a non-selective antagonist of mAChRs, mitigated AChEI-mediated protection. Mice pretreated with mecamylamine had an improved response in glucose tolerance test (GTT) than mice pretreated with atropine. These differential effects of nAChR and mAChR antagonists correlated with the extent of islet cell infiltration and with the structure and functionality of the ß-cells. Taken together, our data suggest that mAChRs are essential for the protective effect of cholinergic stimulation in autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolinesterasa/sangre , Animales , Atropina/farmacología , Diabetes Mellitus Experimental/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/sangre , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Mecamilamina/farmacología , Ratones , Ratones Endogámicos C57BL , Antagonistas Muscarínicos/farmacología , Antagonistas Nicotínicos/farmacología , Paraoxon/farmacología , Paraoxon/uso terapéutico , Estreptozocina/farmacología
9.
Front Oncol ; 7: 167, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28856117

RESUMEN

There is renewed interest in the potential use of natural compounds in cancer therapy. Previously, we demonstrated the anti-tumor properties of manuka honey (MH) against several cancers. However, the underlying mechanism and molecular targets of this activity remain unknown. For this study, the early targets of MH and its modulatory effects on proliferation, invasiveness, and angiogenic potential were investigated using two human breast cancer cell lines, the triple-negative MDA-MB-231 cells and estrogen receptor-positive MCF-7 cells, and the non-neoplastic breast epithelial MCF-10A cell line. Exposure to MH at concentrations of 0.3-1.25% (w/v) induced a dose-dependent inhibition of the proliferation of MDA-MB-231 and MCF-7, but not MCF-10A, cells. This inhibition was independent of the sugar content of MH as a solution containing equivalent concentrations of its three major sugars failed to inhibit cell proliferation. At higher concentrations (>2.5%), MH was found to be generally deleterious to the growth of all three cell lines. MH induced apoptosis of MDA-MB-231 cells through activation of caspases 8, 9, 6, and 3/7 and this correlated with a loss of Bcl-2 and increased Bax protein expression in MH-treated cells. Incubation with MH induced a time-dependent translocation of cytochrome c from mitochondria to the cytosol and Bax translocation from the cytosol into the mitochondria. MH also induced apoptosis of MCF-7 cells via the activation of caspases 9 and 6. Low concentrations of MH (0.03-1.25% w/v) induced a rapid reduction in tyrosine-phosphorylated STAT3 (pY-STAT3) in MDA-MB-231 and MCF-7 cells. Maximum inhibition of pY-STAT3 was observed at 1 h with a loss of >80% and coincided with decreased interleukin-6 (IL-6) production. Moreover, MH inhibited the migration and invasion of MDA-MB-231 cells as well as the angiogenic capacity of human umbilical vein endothelial cells. Our findings identify multiple functional pathways affected by MH in human breast cancer and highlight the IL-6/STAT3 signaling pathway as one of the earliest potential targets in this process.

10.
Respir Res ; 18(1): 136, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28693498

RESUMEN

BACKGROUND: Inhibitors of mTOR, such as sirolimus, have been shown to induce thymus involution and inflammatory lung disease in mice. The latter effect supports the role of this serine/threonine kinase in ameliorating lung inflammation. Other studies have shown sirolimus reduces/delays lung disease associated with various strains of influenza A virus (IAV). Thus, the effects of mTOR inhibitors on influenza infection deserve further studies. METHODS: Here, we examined the changes in lung viral copies, pathology and pulmonary function associated with IAV (A/PR/8/34) infection in mice treated with sirolimus. RESULTS: Body weight loss peaked between days 6-11 post-infection and was more severe in IAV-infected mice that were administered sirolimus as compared to mice that received IAV alone (p = 0.030). Natural log viral gene copies, mean ± SD per mg lung tissue, in IAV-infected mice that were administered sirolimus were 17.31 ± 1.27 on day 4, 19.31 ± 7.46 on day 10, and 0 on day 25. The corresponding number of copies in mice that received IAV alone were 18.56 ± 0.95 on day 4 (p = 0.132), 1.52 ± 1.39 on day 10 (p = 0.008), and 0 on day 25. Lung pathology was evident on days 4, 10, and 25 post infection, with mean ± SD inflammatory score of 9.0 ± 4.5 in IAV-infected mice that were administered sirolimus, as compared to 11.5 ± 4.5 (p = 0.335) in mice received IAV alone (maximum score, 26.0). Impaired lung function was evident in IAV-infected mice on days 4 and 10, as demonstrated by increased airway resistance and decreased compliance. CONCLUSIONS: In this model, the effects of sirolimus on influenza infection included severe weight loss and modified viral replication, respiratory function and lung inflammation. The adverse events associated with sirolimus treatment are consistent with its potent immunosuppressive activity and, thus, preclude its use in IAV infection.


Asunto(s)
Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Pulmón/efectos de los fármacos , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Carga Viral/efectos de los fármacos , Animales , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Gripe Humana/patología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Sirolimus/farmacología , Carga Viral/fisiología
11.
Front Immunol ; 7: 419, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27790217

RESUMEN

Type I diabetes (T1D) results from T cell-mediated damage of pancreatic ß-cells and loss of insulin production. The cholinergic anti-inflammatory pathway represents a physiological link connecting the central nervous and immune systems via vagus nerve, and functions to control the release of proinflammatory cytokines. Using the multiple low-dose streptozotocin (MLD-STZ) model to induce experimental autoimmune diabetes, we investigated the potential of regulating the development of hyperglycemia through administration of paraoxon, a highly specific acetylcholinesterase inhibitor (AChEI). We demonstrate that pretreatment with paraoxon prevented hyperglycemia in STZ-treated C57BL/6 mice. This correlated with a reduction in T cell infiltration into pancreatic islets and preservation of the structure and functionality of ß-cells. Gene expression analysis of pancreatic tissue revealed that increased peripheral cholinergic activity prevented STZ-mediated loss of insulin production, this being associated with a reduction in IL-1ß, IL-6, and IL-17 proinflammatory cytokines. Intracellular cytokine analysis in splenic T cells demonstrated that inhibition of AChE led to a shift in STZ-induced immune response from a predominantly disease-causing IL-17-expressing Th17 cells to IFNγ-positive Th1 cells. Consistent with this conclusion, inhibition of AChE failed to prevent STZ-induced hyperglycemia in IFNγ-deficient mice. Our results provide mechanistic evidence for the prevention of murine T1D by inhibition of AChE and suggest a promising strategy for modulating disease severity.

12.
PLoS One ; 8(2): e55993, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23409104

RESUMEN

Manuka honey has been recognized for its anti-bacterial and wound-healing activity but its potential antitumor effect is poorly studied despite the fact that it contains many antioxidant compounds. In this study, we investigated the antiproliferative activity of manuka honey on three different cancer cell lines, murine melanoma (B16.F1) and colorectal carcinoma (CT26) as well as human breast cancer (MCF-7) cells in vitro. The data demonstrate that manuka honey has potent anti-proliferative effect on all three cancer cell lines in a time- and dose-dependent manner, being effective at concentrations as low as 0.6% (w/v). This effect is mediated via the activation of a caspase 9-dependent apoptotic pathway, leading to the induction of caspase 3, reduced Bcl-2 expression, DNA fragmentation and cell death. Combination treatment of cancer cells with manuka and paclitaxel in vitro, however, revealed no evidence of a synergistic action on cancer cell proliferation. Furthermore, we utilized an in vivo syngeneic mouse melanoma model to assess the potential effect of intravenously-administered manuka honey, alone or in combination with paclitaxel, on the growth of established tumors. Our findings indicate that systemic administration of manuka honey was not associated with any alterations in haematological or clinical chemistry values in serum of treated mice, demonstrating its safety profile. Treatment with manuka honey alone resulted in about 33% inhibition of tumor growth, which correlated with histologically observable increase in tumor apoptosis. Although better control of tumor growth was observed in animals treated with paclitaxel alone or in combination with manuka honey (61% inhibition), a dramatic improvement in host survival was seen in the co-treatment group. This highlights a potentially novel role for manuka honey in alleviating chemotherapy-induced toxicity.


Asunto(s)
Antineoplásicos/farmacología , Miel , Leptospermum/química , Melanoma/tratamiento farmacológico , Administración Intravenosa , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Masculino , Melanoma/mortalidad , Melanoma/patología , Ratones , Necrosis , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Paclitaxel/toxicidad , Carga Tumoral/efectos de los fármacos
13.
Immune Netw ; 11(5): 268-80, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22194710

RESUMEN

BACKGROUND: Dengue virus, which belongs to the Flavivirus genus of the Flaviviridae family, causes fatal dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) with infection risk of 2.5 billion people worldwide. However, approved vaccines are still not available. Here, we explored the immune responses induced by alternating prime-boost vaccination using DNA vaccine, adenovirus, and vaccinia virus expressing E protein of dengue virus type 2 (DenV2). METHODS: Following immunization with DNA vaccine (pDE), adenovirus (rAd-E), and/or vaccinia virus (VV-E) expressing E protein, E protein-specific IgG and its isotypes were determined by conventional ELISA. Intracellular CD154 and cytokine staining was used for enumerating CD4+ T cells specific for E protein. E protein-specific CD8+ T cell responses were evaluated by in vivo CTL killing activity and intracellular IFN-γ staining. RESULTS: Among three constructs, VV-E induced the most potent IgG responses, Th1-type cytokine production by stimulated CD4+ T cells, and the CD8+ T cell response. Furthermore, when the three constructs were used for alternating prime-boost vaccination, the results revealed a different pattern of CD4+ and CD8+ T cell responses. i) Priming with VV-E induced higher E-specific IgG level but it was decreased rapidly. ii) Strong CD8+ T cell responses specific for E protein were induced when VV-E was used for the priming step, and such CD8+ T cell responses were significantly boosted with pDE. iii) Priming with rAd-E induced stronger CD4+ T cell responses which subsequently boosted with pDE to a greater extent than VV-E and rAd-E. CONCLUSION: These results indicate that priming with live viral vector vaccines could induce different patterns of E protein- specific CD4+ and CD8+ T cell responses which were significantly enhanced by booster vaccination with the DNA vaccine. Therefore, our observation will provide valuable information for the establishment of optimal prime-boost vaccination against DenV.

14.
J Immunol ; 185(3): 1429-41, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20581148

RESUMEN

Japanese encephalitis virus (JEV) is a frequent cause of acute and epidemic viral encephalitis. However, there is little information describing the mechanisms by which JEV subverts immune responses that may predispose the host to secondary infections. In this study, we found that JEV induced the subversion of CD8(+) T cell responses in a transient manner that was closely correlated with viral multiplication. Subsequently, analysis using a TCR-transgenic system revealed that CD8(+) T cells purified from JEV-infected mice showed impaired responses, and that naive CD8(+) T cells adoptively transferred into JEV-infected recipients showed less expanded responses. Furthermore, JEV altered the splenic dendritic cell (DC) subpopulation via preferential depletion of CD8alpha(+)CD11c(+) DCs without changing the plasmacytoid DCs and induced a significant reduction in the surface expression of MHC class II and CD40, but not MHC class I, CD80, and CD86 molecules. Finally, JEV was shown to inhibit the presentation of MHC class I-restricted Ag in DCs, and this immune suppression was ameliorated via the activation of DCs by TLR agonists. Collectively, our data indicate that JEV precludes the functions of Ag-presenting machinery, such as depletion of CD8alpha(+)CD11c(+) DCs and downregulation of MHC class I-restricted Ag presentation, thereby leading to immune subversion of CD8(+) T cells.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Evasión Inmune/inmunología , Animales , Antígeno CD11c/biosíntesis , Antígeno CD11c/metabolismo , Antígenos CD8/biosíntesis , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/patología , Chlorocebus aethiops , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/patología , Encefalitis Japonesa/virología , Epítopos de Linfocito T/inmunología , Herpesvirus Humano 1/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Vero
15.
J Immunol ; 183(4): 2462-74, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19635909

RESUMEN

Dendritic cells (DCs) are potent initiators of T cell-mediated immunity that undergo maturation during viral infections. However, few reports describing the interactions of DCs with Japanese encephalitis virus (JEV), which remains the most frequent cause of acute and epidemic viral encephalitis, are available. In this study, we investigated the interaction of JEV with DCs and macrophages. JEV replicated its viral RNA in both cells with different efficiency, and JEV infection of macrophages followed the classical activation pathway of up-regulation of tested costimulatory molecules and proinflammatory cytokine production (IL-6, TNF-alpha, and IL-12). On the contrary, JEV-infected DCs failed to up-regulate costimulatory molecules such as CD40 and MHC class II. Of more interest, along with production of proinflammatory cytokines, DCs infected by JEV released antiinflammatory cytokine IL-10, which was not detected in macrophages. Moreover, signaling through MyD88 molecule, a pan-adaptor molecule of TLRs, and p38 MAPK in JEV-infected DCs was found to play a role in the production of cytokines and subversion of primary CD4(+) and CD8(+) T cell responses. We also found that IL-10 released from JEV-infected DCs led to a reduction in the priming of CD8(+) T cells, but not CD4(+) T cells. Taken together, our data suggest that JEV induces functional impairment of DCs through MyD88-dependent and -independent pathways, which subsequently leads to poor CD4(+) and CD8(+) T cell responses, resulting in boosting viral survival and dissemination in the body.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Macrófagos/inmunología , Macrófagos/virología , Factor 88 de Diferenciación Mieloide/fisiología , Transducción de Señal/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Células Cultivadas , Células Dendríticas/metabolismo , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/patología , Encefalitis Japonesa/virología , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Replicación Viral/inmunología
16.
Immunol Cell Biol ; 87(1): 91-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18794906

RESUMEN

The CC chemokine receptor 7 (CCR7) and cognate CCR7 ligands, CCL19 and CCL21, help establish microenvironments in lymphoid tissue that can facilitate encounters between naive T cells and mature dendritic cells (DCs). This study was conducted to determine if CCR7 ligands can augment the immunogenicity of a DNA vaccine that expresses glycoprotein B (gB) of the pseudorabies virus (PrV). The genetic co-transfer of CCR7 ligands along with a PrV DNA vaccine increased the levels of serum PrV-specific immunoglobulin (Ig) G by 2- to 2.5-fold. In addition, the level of PrV-specific IgG2a isotype was significantly enhanced by co-injection of CCR7 ligand DNA, which indicates that CCR7 ligand biases the humoral immunity toward the Th1-type pattern. The co-injection of CCR7 ligand DNA consistently enhanced the level of Th1-type cytokines (IL-2 and IFN-gamma) produced by stimulated immune cells when compared with a group that was vaccinated with the PrV DNA vaccine. Also, the genetic co-transfer of CCR7 ligand DNAs with PrV DNA vaccine provided prolonged survival against a virulent challenge by PrV. Moreover, the co-administration of CCR7 ligand DNA increased the number of mature DCs into the secondary lymphoid tissues, which appeared to enhance the proliferation of PrV-immune CD4(+) T cells. Taken together, these findings indicate that CCR7 ligands are an attractive adjuvant for a PrV DNA vaccine that can offer protective immunity against the PrV.


Asunto(s)
Quimiocina CCL19/inmunología , Quimiocina CCL21/inmunología , Vacunas contra la Seudorrabia/inmunología , Seudorrabia/prevención & control , Receptores CCR7/inmunología , Vacunas de ADN/inmunología , Animales , Formación de Anticuerpos , Quimiocina CCL19/genética , Quimiocina CCL21/genética , Citocinas/biosíntesis , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Femenino , Ratones , Ratones Endogámicos C57BL , Seudorrabia/inmunología , Vacunas contra la Seudorrabia/administración & dosificación , Vacunas contra la Seudorrabia/genética , Transfección , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Proteínas del Envoltorio Viral/inmunología
17.
J Microbiol Biotechnol ; 18(7): 1326-34, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18667863

RESUMEN

The prime-boost vaccination with DNA vaccine and recombinant viral vector has emerged as an effective prophylactic strategy to control infectious diseases. Here, we compared the protective immunities induced by multiple alternating immunizations with DNA vaccine (pCIgB) and replication-incompetent adenovirus (Ad-gB) expressing glycoprotein gB of pseudorabies virus (PrV). The platform of pCIgB-prime and Ad-gB-boost induced the most effective immune responses and provided protection against virulent PrV infection. However, priming with pCIgB prior to vaccinating animals by the DNA vaccine-prime and Ad-boost protocol provided neither effective immune responses nor protection against PrV. Similarly, boosting with Ad-gB following immunization with DNA vaccine-prime and Ad-boost showed no significant responses. Moreover, whereas the administration of Ad-gB for primary immunization induced Th2-type-biased immunity, priming with pCIgB induced Th1-type-biased immunity, as judged by the production of PrV-specific IgG isotypes and cytokine IFN-gamma. These results indicate that the order and injection frequency of vaccine vehicles used for heterologous prime-boost vaccination affect the magnitude and nature of the immunity. Therefore, our demonstration implies that the prime-boost protocol should be carefully considered and selected to induce the desired immune responses.


Asunto(s)
Adenoviridae/metabolismo , Expresión Génica , Herpesvirus Suido 1/inmunología , Vacunas contra la Seudorrabia/administración & dosificación , Seudorrabia/prevención & control , Vacunación , Vacunas de ADN/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Adenoviridae/genética , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Citocinas/inmunología , Femenino , Herpesvirus Suido 1/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Seudorrabia/inmunología , Seudorrabia/virología , Vacunas contra la Seudorrabia/genética , Vacunas contra la Seudorrabia/inmunología , Vacunación/métodos , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/genética , Replicación Viral
18.
Exp Mol Med ; 40(6): 583-95, 2008 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-19116444

RESUMEN

Replication-incompetent adenoviruses expressing three major glycoproteins (gB, gC, and gD) of pseudorabies virus (PrV) were constructed and used to examine the ability of these glycoproteins to induce protective immunity against a lethal challenge. Among three constructs, recombinant adenovirus expressing gB (rAd-gB) was found to induce the most potent immunity biased to Th1-type, as determined by the IgG isotype ratio and the profile of the Th1/Th2 cytokine production. Conversely, the gC-expressing adenovirus (rAd-gC) revealed Th2-type immunity and the gD-expressing adenovirus (rAd-gD) induced lower levels of IFN-? and IL-4 production than other constructs, except IL-2 production. Mucosal delivery of rAd-gB induced mucosal IgA and serum IgG responses and biased toward Th2-type immune responses. However, these effects were not observed in response to systemic delivery of rAd-gB. In addition, rAd-gB appeared to induce effective protective immunity against a virulent viral infection, regardless of whether it was administered via the muscular or systemic route. These results suggest that administration of replication-incompetent adenoviruses can induce different types of immunity depending on the expressed antigen and that recombinant adenoviruses expressing gB induced the most potent Th1-biased humoral and cellular immunity and provided effective protection against PrV infection.


Asunto(s)
Adenoviridae/inmunología , Glicoproteínas/inmunología , Herpesvirus Suido 1/inmunología , Vacunas contra la Seudorrabia/inmunología , Seudorrabia/inmunología , Replicación Viral , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Formación de Anticuerpos , Línea Celular , Citocinas/inmunología , Femenino , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/fisiología , Inmunidad Celular , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Seudorrabia/prevención & control , Vacunas contra la Seudorrabia/administración & dosificación , Porcinos , Células TH1/inmunología , Células Th2/inmunología
19.
Res Vet Sci ; 83(1): 73-81, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17196231

RESUMEN

To assess the correlation between the nature of immunity induced by different types of immunogens and the establishment of latent infection by wild-type pseudorabies virus (PrV), we used a murine model immunized with different immunogens, the PrV modified live vaccine (MLV), inactivated vaccine (IAV), and commercial oil-adjuvant subunit vaccine (OSV), via either intranasal (i.n.) or intramuscular (i.m.) route. Both MLV and IAV induced a different nature of immunity biased to Th1- and Th2-type, respectively, as judged by the ratio of PrV-specific IgG isotypes (IgG2a/IgG1) and the profile of cytokine IL-2, IL-4, and IFN-gamma production. In contrast, the OSV induced a lower isotype IgG2a to IgG1 ratio and higher level of IL-2 production. The MLV (inducing Th1-type) provided more effective protection against a virulent wild-type PrV challenge than IAV and OSV (inducing Th2- and mixed type, respectively). In addition, the MLV impeded the establishment of a latent infection with wild-type PrV, and the decrease in the PrV latency load by immunization with the MLV appeared to be mediated by the immune T-cells. These results demonstrate the substantial role of the immune responses driven by preceding vaccination in modulating the establishment of PrV latency caused by the post-infection of a field virus.


Asunto(s)
Herpesvirus Suido 1/inmunología , Vacunas contra la Seudorrabia/inmunología , Seudorrabia/prevención & control , Seudorrabia/virología , Latencia del Virus/fisiología , Animales , Anticuerpos Antivirales/sangre , Vías de Administración de Medicamentos , Femenino , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos C57BL , Seudorrabia/inmunología , Vacunas contra la Seudorrabia/administración & dosificación
20.
J Microbiol Biotechnol ; 17(12): 1955-64, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18167442

RESUMEN

A recent report showed that analysis of CD154 expression in the presence of the secretion inhibitor Brefeldin A (Bref A) could be used to assess the entire repertoire of antigen-specific CD4(+) T helper cells. However, the capacity of intracellular CD154 expression to identify antigen-specific CD8(+) T cells has yet to be investigated. In this study, we compared the ability of intracellular CD154 expression to assess antigen-specific CD8(+) T cells with that of accepted standard assays, namely intracellular cytokine IFN-gamma staining (ICS) and MHC class I tetramer staining. The detection of intracellular CD154 molecules in the presence of Bref A reflected the kinetic trend of antigen-specific CD8+ T cell number, but unfortunately showed less sensitivity than ICS and tetramer staining. However, ICS levels peaked and saturated 8 h after antigenic stimulation in the presence of Bref A and then declined, whereas intracellular CD154 expression peaked by 8 h and maintained the saturated level up to 24 h post-stimulation. Moreover, intracellular CD154 expression in antigen-specific CD8+ T cells developed in the absence of CD4(+) T cells changed little, whereas the number of IFN-gamma-producing CD8(+) T cells decreased abruptly. These results suggest that intracellular CD154 could aid the assessment of antigen-specific CD8(+) T cells, but does not have as much ability to identify heterogeneous CD4(+) T helper cells. Therefore, the combined analytical techniques of ICS and tetramer staining together with intracellular CD154 assays may be able to provide useful information on the accurate phenotype and functionality of antigen-specific CD8(+) T cells.


Asunto(s)
Ligando de CD40/biosíntesis , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Interferón gamma/biosíntesis , Traslado Adoptivo , Animales , Antivirales/farmacología , Brefeldino A/farmacología , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/inmunología , Proteínas del Huevo/inmunología , Proteínas del Huevo/farmacología , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/inmunología , Interferón gamma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Fragmentos de Péptidos , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...