全文获取类型
收费全文 | 81篇 |
免费 | 7篇 |
专业分类
88篇 |
出版年
2024年 | 1篇 |
2022年 | 3篇 |
2021年 | 6篇 |
2020年 | 2篇 |
2019年 | 3篇 |
2018年 | 4篇 |
2017年 | 2篇 |
2016年 | 3篇 |
2015年 | 1篇 |
2014年 | 5篇 |
2013年 | 11篇 |
2012年 | 9篇 |
2011年 | 9篇 |
2010年 | 2篇 |
2009年 | 2篇 |
2008年 | 4篇 |
2007年 | 2篇 |
2006年 | 2篇 |
2005年 | 1篇 |
2004年 | 2篇 |
2002年 | 2篇 |
2001年 | 4篇 |
2000年 | 1篇 |
1999年 | 1篇 |
1998年 | 1篇 |
1997年 | 1篇 |
1995年 | 1篇 |
1985年 | 1篇 |
1981年 | 1篇 |
1980年 | 1篇 |
排序方式: 共有88条查询结果,搜索用时 31 毫秒
61.
Cody McHale Zahraa Mohammed Juline Deppen Gregorio Gomez 《Biochimica et Biophysica Acta (BBA)/General Subjects》2018,1862(5):1069-1078
Background
Interleukin-6 is a gp130 utilizing cytokine that is consistently associated with allergic diseases like asthma and urticaria in humans where mast cells are known to play a critical role. However, the role of IL-6 in allergic disease in not known. IL-6 was reported to enhance degranulation of in vitro-derived mast cells, but the effect of IL-6 on mediator release from human in situ-matured tissue-isolated mast cells had not been reported.Methods
Human mature mast cells were isolated and purified from normal skin tissue from different donors. The expression of surface-expressed IL-6 receptors was demonstrated by flow cytometry. The effect of IL-6 on FcεRI-induced degranulation, PGD2 biosynthesis, and cytokine production was determined with β?hexosaminidase release assay, Western blotting, quantitative real-time PCR, and ELISA. The small molecule inhibitor of STAT-3, C188-9, was used to demonstrate STAT3 dependency.Results: IL-6 significantly potentiated FcεRI-induced PGD2 biosynthesis, but had no effect on degranulation. IL-6 also induced VEGF gene expression and protein secretion, and enhanced FcεRI-induced IL-8 production. Mechanistically, IL-6 enhanced FcεRI-induced COX?2 expression, PGD2 biosynthesis, and VEGF production in a STAT3 dependent manner.Conclusion
Here, we demonstrate that IL-6 is a potentiator of FcεRI-induced PGD2 biosynthesis, and can induce or enhance production of pro-angiogenesis factors VEGF and IL-8 from human in situ-matured skin mast cells.General significance
These findings from this study indicate that IL-6 contributes to human allergic disease by enhancing the production of inflammatory PGD2 from tissue-resident mast cells. Moreover, the data suggest a novel role for IL-6 in mast cell-mediated angiogenesis. 相似文献62.
63.
Blanche P. Alter Sabra C. Goff Thomas J. Klonowski Michael D. Garrick 《Preparative biochemistry & biotechnology》2013,43(5):633-644
Polyacrylamide gel electrophoresis in urea and Triton X-100 of a hemolysate from human fetal red blood cells produces four major protein bands: α, β, and 2 γ globin chains. We have verified that the latter two are the Gγ and Aγ globin chains which have respectively glycine or alanine at position 136. After incorporation of either [3H] alanine or [3H] glycine into newly synthesized globin each y chain was isolated by preparative electrophoresis. The chains were cleaved with cyanogen bromide at methionines 55 and 133, then subjected to automated sequencing, and the residues from each sequencer turn counted. Glycine incorporation was detected for the third turn (position 136) of the Gγ chain and alanine for the Aγ Substantial metabolic conversion of [3H] glycine to serine and proline was also noted. 相似文献
64.
65.
Hantaviruses, similar to several emerging zoonotic viruses, persistently infect their natural reservoir hosts, without causing overt signs of disease. Spillover to incidental human hosts results in morbidity and mortality mediated by excessive proinflammatory and cellular immune responses. The mechanisms mediating the persistence of hantaviruses and the absence of clinical symptoms in rodent reservoirs are only starting to be uncovered. Recent studies indicate that during hantavirus infection, proinflammatory and antiviral responses are reduced and regulatory responses are elevated at sites of increased virus replication in rodents. The recent discovery of structural and non-structural proteins that suppress type I interferon responses in humans suggests that immune responses in rodent hosts could be mediated directly by the virus. Alternatively, several host factors, including sex steroids, glucocorticoids, and genetic factors, are reported to alter host susceptibility and may contribute to persistence of hantaviruses in rodents. Humans and reservoir hosts differ in infection outcomes and in immune responses to hantavirus infection; thus, understanding the mechanisms mediating viral persistence and the absence of disease in rodents may provide insight into the prevention and treatment of disease in humans. Consideration of the coevolutionary mechanisms mediating hantaviral persistence and rodent host survival is providing insight into the mechanisms by which zoonotic viruses have remained in the environment for millions of years and continue to be transmitted to humans.Hantaviruses are negative sense, enveloped RNA viruses (family: Bunyaviridae) that are comprised of three RNA segments, designated small (S), medium (M), and large (L), which encode the viral nucleocapsid (N), envelope glycoproteins (GN and GC), and an RNA polymerase (Pol), respectively. More than 50 hantaviruses have been found worldwide [1]. Each hantavirus appears to have coevolved with a specific rodent or insectivore host as similar phylogenetic trees are produced from virus and host mitochondrial gene sequences [2]. Spillover to humans causes hemorrhagic fever with renal syndrome (HFRS) or hantavirus cardiopulmonary syndrome (HCPS), depending on the virus [3]–[5]. Although symptoms vary, a common feature of both HFRS and HCPS is increased permeability of the vasculature and mononuclear infiltration [4]. Pathogenesis of HRFS and HCPS in humans is hypothesized to be mediated by excessive proinflammatory and CD8+ T cell responses ().
Open in a separate windowaSNV, Sin Nombre virus; NY-1V, New York-1 virus; PUUV, Puumala virus; PHV, Prospect Hill virus; ANDV, Andes virus; TULV, Tula virus; HTNV, Hantaan virus; DOBV, Dobrava virus.bHUVEC, human umbilical vascular endothelial cells; HSVEC, human saphenous vein endothelial cells; HMVEC-L, human lung microvascular endothelial cells; COS-7, African green monkey kidney fibroblasts transformed with Simian virus 40; MRC5, human fetal lung fibroblasts; MФ, macrophages; DCs, dendritic cells; BAL, bronchoalveolar lavage, PBMC, human peripheral blood mononuclear cells.cAcute infection is during symptomatic disease in patients.dSuppressor T cells likely represent cells currently referred to as regulatory T cells.
Open in a separate windowaSEOV, Seoul virus; HTNV, Hantaan virus, PUUV, Puumala virus; SNV, Sin Nombre virus; PUUV, Puumala virus; BCCV, Black Creek Canal virus.bMФ, macrophages.cAcute infection is <30 days p.i. and persistent infection is ≥30 days p.i.d
Mus musculus, non-natural reservoir host for hantaviruses.In contrast to humans, hantaviruses persistently infect their reservoir hosts, presumably causing lifelong infections [6]. Hantaviruses are shed in saliva, urine, and feces, and transmission among rodents or from rodents to humans occurs by inhalation of aerosolized virus in excrement or by transmission of virus in saliva during wounding [7],[8]. Although widely disseminated throughout the rodent host, high amounts of hantaviral RNA and antigen are consistently identified in the lungs of their rodent hosts, suggesting that the lungs may be an important site for maintenance of hantaviruses during persistent infection [9]–[18]. Hantavirus infection in rodents is characterized by an acute phase of peak viremia, viral shedding, and virus replication in target tissues, followed by a persistent phase of reduced, cyclical virus replication despite the presence of high antibody titers (Figure 1) [12]–[16], [18]–[20]. The onset of persistent infection varies across hantavirus–rodent systems, but generally the acute phase occurs during the first 2–3 weeks of infection and virus persistence is established thereafter (Figure 1).Open in a separate windowFigure 1Kinetics of Hantavirus Infection in Rodents.Adapted from Lee et al. [15] and others [12]–[14],[16],[18],[20], the kinetics of relative hantaviral load in blood (red), saliva (green), and lung tissue (blue) and antibody responses (black) during the acute and persistent phases of infection are represented. The amount of genomic viral RNA, infectious virus titer, and/or relative amount of viral antigen have been incorporated as relative hantaviral load. The antibody response is integrated as the relative amount of anti-hantavirus IgG and/or neutralizing antibody titers.Hantavirus infection alone does not cause disease, as reservoir hosts and non-natural hosts (e.g., hamsters infected with Sin Nombre virus [SNV] or Choclo virus) may support replicating virus in the absence of overt disease [12],[14],[16],[18],[21],[22]. Our primary hypothesis is that certain immune responses that are mounted in humans during hantavirus infection are suppressed in rodent reservoirs to establish and maintain viral persistence, while preventing disease (相似文献
Table 1
Summary of Immune Responses in Humans during Hantavirus Infection.Categorical Response | Immune Marker | Effect of Infection | Virus Speciesa | In Vitro/In Vivo | Tissue or Cell Typeb, Phase of Infectionc | References |
Innate | RIG-I | Elevated | SNV | In vitro | HUVEC, ≤24 h p.i. | [79] |
Reduced | NY-1V | In vitro | HUVEC, ≤24 h p.i. | [37] | ||
TLR3 | Elevated | SNV | In vitro | HUVEC, ≤24 h p.i. | [79] | |
IFN-β | Elevated | PUUV, PHV, ANDV | In vitro | HSVEC, HMVEC-L, ≤24 h p.i. | [36],[80] | |
Reduced | TULV, PUUV NSs | In vitro | COS-7 and MRC5 cells, ≤24 h p.i. | [32],[33] | ||
IFN-α | Elevated | PUUV, HTNV | In vitro | MФ, DCs, 4 days p.i. | [30] | |
No change | HTNV | In vivo | Blood, acute | [81] | ||
IRF-3, IRF-7 | Elevated | SNV, HTNV, PHV, ANDV | In vitro | HMVEC-L, ≤24 h p.i. | [33],[38] | |
MxA | Elevated | HTNV, NY-1V, PHV, PUUV, ANDV, SNV, TULV | In vitro | MФ,HUVEC,HMVEC-L, 6 h–4 days p.i. | [36], [39]–[41],[79] | |
MHC I and II | Elevated | HTNV | In vitro | DCs, 4 days p.i. | [30] | |
CD11b | Elevated | PUUV | In vivo | Blood, acute | [82] | |
CD40, CD80, CD86 | Elevated | HTNV | In vitro | DCs, 4 days p.i. | [30],[83] | |
NK cells | Elevated | PUUV | In vivo | BAL, acute | [84] | |
Proinflammatory/Adhesion | IL-1β | Elevated | SNV, HTNV | In vivo | Blood, lungs, acute | [85],[86] |
IL-6 | Elevated | SNV, PUUV | In vivo | Blood, lungs, acute | [85],[87],[88] | |
TNF-α | Elevated | PUUV, SNV, HTNV | In vivo | Blood, lungs, kidney, acute | [85],[86],[88],[89] | |
Elevated | HTNV | In vitro | DCs, 4 days p.i. | [30] | ||
CCL5 | Elevated | SNV, HTNV | In vitro | HMVEC-L, HUVEC, 12 h–4 days p.i. | [38],[39],[90] | |
CXCL8 | Elevated | PUUV | In vivo | Blood, acute | [82] | |
Elevated | PUUV | In vivo | Men, blood, acute | [62] | ||
Elevated | TULV, PHV, HTNV | In vitro | HUVEC, MФ, 2–4 days p.i. | [39],[91] | ||
CXCL10 | Elevated | SNV, HTNV, PHV | In vitro | HMVEC-L,HUVEC, 3–4 days p.i. | [38],[39] | |
Elevated | PUUV | In vivo | Men, blood, acute | [62] | ||
IL-2 | Elevated | SNV, HTNV, PUUV | In vivo | Blood, lungs, acute | [82],[86] | |
Nitric oxide | Elevated | PUUV | In vivo | Blood, acute | [92] | |
GM-CSF | Elevated | PUUV | In vivo | Women, blood, acute | [62] | |
ICAM, VCAM | Elevated | PUUV | In vivo | Kidney, acute | [87] | |
Elevated | HTNV, PHV | In vitro | HUVEC, 3–4 days p.i. | [30],[39] | ||
E-selectin | Elevated | PUUV | In vivo | Blood, acute | [82] | |
CD8+ and CD4+ T cells | IFN-γ | Elevated | HTNV, SNV | In vivo | Blood, CD4+,CD8+, lungs, acute | [81],[86] |
CD8+ | Elevated | DOBV, PUUV, HTNV | In vivo | Blood, BAL, acute | [52],[84],[93] | |
Virus-specific IFN-γ+CD8+ | Elevated | PUUV, SNV | In vivo | PBMC, acute | [45],[94] | |
Perforin, Granzyme B | Elevated | PUUV | In vivo | Blood, acute | [95] | |
CD4+CD25+ “activated” | Elevated | DOBV, PUUV | In vivo | PBMC, acute | [89],[93] | |
IL-4 | Elevated | SNV | In vivo | Lungs, acute | [86] | |
Regulatory | “suppressor T cells”d | Reduced | HTNV | In vivo | Blood, acute | [52] |
IL-10 | Elevated | PUUV | In vivo | Blood, acute | [86] | |
TGF-β | Elevated | PUUV | In vivo | Kidney, acute | [89] | |
Humoral | IgM, IgG, IgA, IgE | Elevated | All hantaviruses | In vivo | Blood | [4] |
Table 2
Summary of Immune Responses in Rodents during Hantavirus Infection.Categorical Response | Immune Marker | Effect of Infection | Virus Speciesa | Host, Tissue or Cell Typeb | Phase of Infectionc | References |
Innate | TLR7 | Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [19] |
Elevated | SEOV | Female Norway rats, lungs | Acute, Persistent | [19] | ||
RIG-I | Elevated | SEOV | Female Norway rats, lungs | Acute, Persistent | [19] | |
Elevated | SEOV | Newborn rats, thalamus | Acute | [96] | ||
TLR3 | Elevated | SEOV | Male Norway rats, lungs | Acute, Persistent | [19] | |
IFN-β | Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [19],[61] | |
Elevated | SEOV | Female Norway rat lungs | Acute | [19],[61] | ||
Mx2 | Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [19],[60] | |
Elevated | SEOV | Female Norway rats, lungs | Acute, Persistent | [19],[60] | ||
Elevated | HTNV, SEOV | Miced, fibroblasts transfected with Mx2 | 3–4 days p.i. | [97] | ||
JAK2 | Elevated | SEOV | Female Norway rats, lungs | Acute | [60] | |
MHC II | Elevated | PUUV | Bank voles | Genetic susceptibility | [74] | |
Proinflammatory/Adhesion | IL-1β | Reduced | SEOV | Male Norway rats, lungs | Persistent | [29] |
IL-6 | Reduced | SEOV | Male and female Norway rats, lungs | Acute, Persistent | [29],[61] | |
Elevated | SEOV | Male rats, spleen | Acute | [29] | ||
TNF-α | Reduced | HTNV | Newborn miced, CD8+, spleen | Acute | [49],[50] | |
Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [29],[42],[61] | ||
Elevated | SEOV | Female Norway rats, lungs | Persistent | [61] | ||
CX3CL1, CXCL10 | Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [29] | |
Elevated | SEOV | Male Norway rats, spleen | Acute | [29] | ||
CCL2, CCL5 | Elevated | SEOV | Male Norway rats, spleen | Acute | [29] | |
NOS2 | Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [29],[61] | |
Elevated | SEOV | Male Norway rats, spleen | Acute | [29] | ||
Elevated | HTNV | Mouse MФd, in vitro | 6 h p.i. | [98] | ||
VCAM, VEGF | Elevated | SEOV | Male Norway rats, spleen | Acute | [29] | |
CD8+ and CD4+ T cells | CD8+ | Reduced | HTNV | Newborn miced, spleen | Persistent | [50] |
Elevated | HTNV | SCID miced, CD8+ transferred, spleen | Persistence | [49] | ||
Elevated | SEOV | Female Norway rats, lungs | Persistent | [61] | ||
IFN-γ | Elevated | SEOV | Female Norway rats, lungs | Persistent | [61] | |
Elevated | SEOV | Male Norway rats, spleen | Acute | [29] | ||
Elevated | SEOV | Male and female Norway rats, splenocytes | Acute | [20] | ||
Elevated | SNV | Deer mice, CD4+ T cells | Acute | [48] | ||
Elevated | HTNV | Newborn miced, CD8+ T cells, spleen | Acute | [50] | ||
Reduced | HTNV | Newborn miced, CD8+ T cells, spleen | Persistent | [99] | ||
IFN-γR | Elevated | SEOV | Female Norway rats, lungs | Acute, Persistent | [60] | |
Reduced | SEOV | Male Norway rats, lungs | Persistent | [60] | ||
T cells | Elevated | SEOV | Nude rats | Persistence | [47] | |
Elevated | HTNV | Nude miced | Persistence | [100] | ||
IL-4 | Reduced | SEOV | Male Norway rats, lungs | Acute, Persistent | [61] | |
Elevated | SNV | Deer mice, CD4+ T cells | Acute | [48] | ||
Elevated | SEOV | Male and female Norway rats, splenocytes | Acute | [20] | ||
Regulatory | Regulatory T cells | Elevated | SEOV | Male Norway rats, lungs | Persistent | [42],[61] |
FoxP3 | Elevated | SEOV | Male Norway rats, lungs | Persistent | [29],[42],[61] | |
TGF-β | Elevated | SEOV | Male Norway rats, lungs | Persistent | [29] | |
SNV | Deer mice, CD4+ T cells | Persistent | [48] | |||
IL-10 | Reduced | SEOV | Male Norway rats, lungs and spleen | Acute, Persistent | [29] | |
Elevated | SNV | Deer mice, CD4+ T cells | Acute | [48] | ||
Humoral | IgG | Elevated | SNV | Deer mice | Persistent | [12],[57] |
Elevated | SEOV | Norway rats | Persistent | [16],[17] | ||
Elevated | HTNV | Field mice | Persistent | [15] | ||
Elevated | PUUV | Bank voles | Persistent | [14] | ||
Elevated | BCCV | Cotton rats | Persistent | [18],[58] |
66.
Blanche P. Alter Sabra C. Goff Thomas J. Klonowski Michael D. Garrick 《Preparative biochemistry & biotechnology》2013,43(2):217-228
Polyacrylamide gel electrophoresis in urea and Triton X-100 of a hemolysate from human fetal red blood cells produces four major protein bands: α, β, and 2 γ globin chains. We have verified that the latter two are the Gγ and Aγ globin chains which have respectively glycine or alanine at position 136. After incorporation of either [3H] alanine or [3H] glycine into newly synthesized globin each y chain was isolated by preparative electrophoresis. The chains were cleaved with cyanogen bromide at methionines 55 and 133, then subjected to automated sequencing, and the residues from each sequencer turn counted. Glycine incor-poration was detected for the third turn (position 136) of the Gγ chain and alanine for the Aγ. Substantial metabolic conversion of [3H] glycine to serine and proline was also noted. 相似文献
67.
Fatma AlZahraa N. AlShahed Hala H. Shoeb Mohammad M. ElShawwa 《Journal of cellular and molecular medicine》2022,26(20):5213
Isotretinoin is an oral retinoid which used across the world in the treatment of patients especially adolescents complaining of acne. In spite of the prevalent clinical use of isotretinoin, the generation of oxidative stress with the affection of several organs leads to the limitation and restriction of its use. Omega‐3 (N‐3) is an essential polyunsaturated fatty acid (PUFAs) with powerful antioxidant properties. The aim of this study was to investigate the histological and biochemical changes occurring in the rat testis following isotretinoin intake and to evaluate the role of omega 3 supplementation in ameliorating testicular damage. Thirty adult male albino rats were divided equally into three groups. Group I is the control group, group II received isotretinoin (1.0 mg/kg/day) dissolved in distilled water and group III received isotretinoin (1.0 mg/kg/day) and omega 3 (400 mg/kg/day). Testis samples were collected and processed for light and electron microscopic examination. The blood samples were collected for biochemical assessments. Results indicated that isotretinoin caused histological changes in all stages of spermatogenesis and alterations of the hormonal assay. These changes in the rat testis which were corrected by omega 3 use. 相似文献
68.
Victor Sebastian-Perez Alfonso García-Rubia Sayed H. Seif el-Din Abdel-Nasser A. Sabra Naglaa M. El-Lakkany Samia William 《Journal of enzyme inhibition and medicinal chemistry》2013,28(1):511-523
Abstract A previous phenotypic screening campaign led to the identification of a quinazoline derivative with promising in vitro activity against Schistosoma mansoni. Follow-up studies of the antischistosomal potential of this candidate are presented here. The in vivo studies in a S. mansoni mouse model show a significant reduction of total worms and a complete disappearance of immature eggs when administered concomitantly with praziquantel in comparison with the administration of praziquantel alone. This fact is of utmost importance because eggs are responsible for the pathology and transmission of the disease. Subsequently, the chemical optimisation of the structure in order to improve the metabolic stability of the parent compound was carried out leading to derivatives with improved drug-like properties. Additionally, the putative target of this new class of antischistosomal compounds was envisaged by using computational tools and the binding mode to the target enzyme, aldose reductase, was proposed. 相似文献
69.
W. Sabra A. M. Haddad A.-P. Zeng 《World journal of microbiology & biotechnology》2014,30(3):1027-1036
Small-colony variants (SCVs) of Pseudomonas aeruginosa are often found in chronically infected airways of patients suffering from cystic fibrosis. These slow-growing morphological variants have been associated with persistent and antibiotic-resistant infections. Nevertheless, the behavior of SCVs under varied availability of O2 and iron, two key variables relevant to the lung environment of CF patients and pathogenicity of P. aeruginosa, has not been systematically studied so far. In this work, the effects of O2 and iron were comparatively studied for a CF P. aeruginosa wild type (WT) strain and its SCV phenotype in a real-time controlled cultivation system. Significant differences in the behavior of these strains were observed and quantified. In general, SCV exhibited a higher fitness than the WT toward aerobic conditions. Under iron rich condition, and despite less release of total extracellular proteins, absence of flagellin and lower siderophore production, the SCV cells grown at fully aerobic conditions showed a higher specific growth rate and a significantly higher cytotoxicity in comparison with the WT cells. The strains behaved also differently towards iron limitation. The phenomena of limited O2 transfer from the gas to the liquid phase and enhancement of formation of virulence factors under conditions of iron limitation were much more profound in the SCV culture than in the WT culture. These results have important implications for better understanding the pathogenicity of P. aeruginosa and its small-colony variants. 相似文献
70.
Shkilnyy A Dubois J Sabra G Sharp J Gagnon S Proulx P Vermette P 《Biotechnology and bioengineering》2012,109(5):1305-1313
This paper reports the optimization of a perfusion bioreactor system previously reported by us (Chouinard et al., 2009). The implementation of a proportional-integral (PI) controller algorithm to control oxygen concentration and pH is presented and discussed. P and I values used by the controller were first estimated using a First-Order-Plus-Dead-Time (FOPDT, Matlab Simulink) and then tuned manually. A new gas exchanger design compatible with the PI controller was introduced and validated to decrease interaction between the injected gases and overall inertia of the system. The gas exchanger was used to adjust both pH and dissolved oxygen (DO) concentration. This new bioreactor system allowed real-time PI control over pH and DO concentration at different flow rates (from 2 to 70 mL min(-1)). Cell viability and proliferation were investigated to validate the updated bioreactor design and performance. 相似文献