首页 | 本学科首页   官方微博 | 高级检索  
文章检索
  按 检索   检索词:      
出版年份:   被引次数:   他引次数: 提示:输入*表示无穷大
  收费全文   2546篇
  免费   152篇
  国内免费   22篇
耳鼻咽喉   9篇
儿科学   59篇
妇产科学   28篇
基础医学   215篇
口腔科学   92篇
临床医学   470篇
内科学   492篇
皮肤病学   31篇
神经病学   299篇
特种医学   205篇
外国民族医学   1篇
外科学   244篇
综合类   63篇
一般理论   1篇
预防医学   186篇
眼科学   32篇
药学   186篇
中国医学   2篇
肿瘤学   105篇
  2021年   33篇
  2019年   30篇
  2018年   29篇
  2017年   28篇
  2016年   37篇
  2015年   30篇
  2014年   45篇
  2013年   102篇
  2012年   93篇
  2011年   121篇
  2010年   58篇
  2009年   91篇
  2008年   85篇
  2007年   92篇
  2006年   82篇
  2005年   92篇
  2004年   58篇
  2003年   89篇
  2002年   68篇
  2001年   69篇
  2000年   75篇
  1999年   54篇
  1998年   55篇
  1997年   52篇
  1996年   64篇
  1995年   44篇
  1994年   31篇
  1993年   35篇
  1992年   46篇
  1991年   45篇
  1990年   54篇
  1989年   65篇
  1988年   54篇
  1987年   57篇
  1986年   35篇
  1985年   51篇
  1984年   40篇
  1983年   38篇
  1982年   37篇
  1981年   28篇
  1980年   33篇
  1979年   35篇
  1978年   32篇
  1977年   28篇
  1976年   25篇
  1974年   22篇
  1972年   27篇
  1968年   23篇
  1967年   22篇
  1966年   25篇
排序方式: 共有2720条查询结果,搜索用时 180 毫秒
61.
Simon  SI; Rochon  YP; Lynam  EB; Smith  CW; Anderson  DC; Sklar  LA 《Blood》1993,82(4):1097-1106
We have recently found that antibodies to L-selectin, the homing receptor on neutrophils, are as effective as those to beta 2-integrin at blocking formyl peptide-stimulated aggregation. Therefore, we investigated the requirements for expression of L-selectin and beta 2- integrin on adjacent cells during aggregation. Fluorescence flow cytometry allowed characterization of aggregates on the basis of size and color, as well as antibody binding to these two adhesive molecules. Formyl peptide-stimulated aggregate formation was measured for individual populations fluorescently labeled red (LDS-751) or green (CD44-FITC), and interpopulation red-green cell conjugates. Blocking either the beta 2-integrin or L-selectin adhesive epitope with monoclonal antibody on individual cell populations resulted in an approximately 50% reduction in two-color aggregation as compared with that in unblocked samples. Shedding the L-selectin on a cell population by preincubation with complexes of lipopolysaccharide and its plasma membrane binding protein also decreased aggregation to a control population by approximately 50%. We examined the aggregation of neutrophils from patients genetically deficient in beta 2-integrin and clinically leukocyte adhesion deficient (LAD). LAD adhesion to normal neutrophils was dependent on the expression of L-selectin on LAD cells and beta 2-integrin on normal cells. Thus, the minimum requirement for adhesion between two mixed populations of neutrophils was that one population expressed the beta 2-integrin and the other expressed the L- selectin adhesive epitope.  相似文献   
62.
To see whether lung transvascular protein sieving occurred during the period of increased microvascular permeability in response to Pseudomonas bacteremia, we measured lung lymph flow and concentrations of eight protein fractions with molecular radii from 36–96 Å during steady state base line periods and during periods of steady state increased permeability after Pseudomonas infusion in five unanesthetized sheep. Lymph clearance (lymph flow × lymph-plasma concentration ratio) for all of the proteins was markedly increased after Pseudomonas, but, as during base line, clearance still decreased with increasing molecular size. We conclude that Pseudomonas bacteremia causes a marked increase in filtration through lung exchanging vessels. The changes in vessel walls result in decreasing passage of protein molecules with increasing molecular size from 36–96 Å.  相似文献   
63.
OBJECTIVE: The aim of this study was to determine the efficacy of epoetin alfa in alleviating anemia and minimizing ribavirin (RBV) dose reductions in patients with chronic hepatitis C virus (HCV) infection receiving combination RBV/interferon alfa (IFN) therapy. METHODS: HCV-infected patients who had Hb levels of 12 g/dl or less during the first 24 wk of combination RBV/IFN therapy (n=64) were randomized to treatment with epoetin alfa (40,000 units) s.c. q.w. or to standard of care (SOC) for anemia management (RBV dose reduction or discontinuation, transfusions). Primary and secondary efficacy endpoints were changes in Hb level and RBV dosage, respectively, from baseline to week 16 of epoetin alfa therapy.Based on intent-to-treat analysis, the mean changes from baseline Hb levels at week 16 were +2.8 g/dl for epoetin alfa versus +0.4 g/dl for SOC (p<0.0001), and the mean changes in RBV dosage were -34 mg/day for epoetin alfa versus -146 mg/day (p=0.060) for SOC. The mean Hb level at week 16 in the epoetin alfa group (13.8 g/dl) was significantly (p<0.0001) higher than that of the SOC group (11.4 g/dl). At week 4 and subsequently, significantly more patients in the epoetin alfa group did not have RBV dosage reductions (p<0.011). At study end, 83% of epoetin alfa-treated patients maintained RBV dosages of at least 800 mg/day, compared with 54% of patients receiving SOC (p=0.022). Epoetin alfa was well tolerated. CONCLUSIONS: In anemic HCV-infected patients treated with RBV/IFN, epoetin alfa increases Hb levels and maintains RBV dosing. Based on these results, epoetin alfa seems to be promising in the treatment of HCV treatment-related anemia. Further research is warranted to determine the potential impact on outcomes, including quality of life and sustained viral response rate.  相似文献   
64.
65.
66.
67.
68.
69.
70.
Neuroinflammation, through production of proinflammatory molecules and activated glial cells, is implicated in Alzheimer''s disease (AD) pathogenesis. One such proinflammatory mediator is tumor necrosis factor α (TNF-α), a multifunctional cytokine produced in excess and associated with amyloid β–driven inflammation and cognitive decline. Long-term global inhibition of TNF receptor type I (TNF-RI) and TNF-RII signaling without cell or stage specificity in triple-transgenic AD mice exacerbates hallmark amyloid and neurofibrillary tangle pathology. These observations revealed that long-term pan anti–TNF-α inhibition accelerates disease, cautions against long-term use of anti–TNF-α therapeutics for AD, and urges more selective regulation of TNF signaling. We used adeno-associated virus vector–delivered siRNAs to selectively knock down neuronal TNF-R signaling. We demonstrate divergent roles for neuronal TNF-RI and TNF-RII where loss of opposing TNF-RII leads to TNF-RI–mediated exacerbation of amyloid β and Tau pathology in aged triple-transgenic AD mice. Dampening of TNF-RII or TNF-RI+RII leads to a stage-independent increase in Iba-1–positive microglial staining, implying that neuronal TNF-RII may act nonautonomously on the microglial cell population. These results reveal that TNF-R signaling is complex, and it is unlikely that all cells and both receptors will respond positively to broad anti–TNF-α treatments at various stages of disease. In aggregate, these data further support the development of cell-, stage-, and/or receptor-specific anti–TNF-α therapeutics for AD.CME Accreditation Statement: This activity (“ASIP 2013 AJP CME Program in Pathogenesis”) has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education (ACCME) through the joint sponsorship of the American Society for Clinical Pathology (ASCP) and the American Society for Investigative Pathology (ASIP). ASCP is accredited by the ACCME to provide continuing medical education for physicians.The ASCP designates this journal-based CME activity (“ASIP 2013 AJP CME Program in Pathogenesis”) for a maximum of 48 AMA PRA Category 1 Credit(s)™. Physicians should only claim credit commensurate with the extent of their participation in the activity.CME Disclosures: The authors of this article and the planning committee members and staff have no relevant financial relationships with commercial interests to disclose.Alzheimer''s disease (AD) pathophysiology is described by chronic and progressive neurodegeneration involving the genesis of extracellular amyloid β (Aβ) plaques, intraneuronal filamentous inclusions called neurofibrillary tangles (NFTs), synapse loss, inflammation, and neuronal cell death, ultimately leading to severe memory loss and cognitive impairment. Neuroinflammation is a highly enigmatic process contributing to disease pathogenesis in AD, where elevated levels of proinflammatory molecules have been associated with Aβ-induced inflammation, neurotoxicity, and cognitive decline.1–4 In AD-afflicted brains, microglia intimately co-localize with Aβ plaques and serve as major sources of proinflammatory mediators, including cytokines and chemokines.5 The pleiotropic proinflammatory cytokine tumor necrosis factor α (TNF-α) is produced in excess concurrently with increased Aβ plaque deposition, an observation that suggests that TNF-α levels reflect the pathologic progression of AD.6–8 Moreover, three TNF-α promoter polymorphisms have been associated with late-onset AD, and two of the three polymorphisms are linked to increased TNF-α production, further connecting this cytokine to the exacerbated chronic inflammatory disease status in AD.9 We and others have demonstrated that TNF-α expression is enhanced in AD mouse models where TNF-α is prepathologically up-regulated in 6-month-old triple-transgenic AD (3xTg-AD) mice,10,11 which corresponds with an enhancement of F4/80-positive microglial cell numbers.12 In addition, when neuron-specific TNF-α is chronically overexpressed in 3xTg-AD mice using adeno-associated virus (AAV) vectors, there is increased severity of inflammation, intracellular Aβ, and Tau pathology that leads to neuronal cell death portending that excessive and unopposed TNF-α signaling enhances AD-associated pathology and is detrimental to neuronal viability.13TNF-α signals through two cognate transmembrane receptors, TNF receptor type I (TNF-RI) and TNF-RII, which are differentially expressed and regulated. TNF-RI is expressed constitutively on most cell types, whereas TNF-RII expression is induced and is restricted to specific cell populations, including hematopoietic cells, microglia, neurons, and endothelial cells.14,15 TNF-R engagement to its ligand mediates distinct cellular responses through the activation of several downstream signal transduction cascades involving the NFκB and JNK pathways. In the context of AD, several reports demonstrate differential roles and activation of TNF-RI and TNF-RII such that genetic deletion of TNF-RI, but not TNF-RII, results in reduced plaque deposition in the APP23 mouse model.16 Moreover, in human brain tissue, TNF-RI protein levels are increased, whereas TNF-RII levels are reduced in patients with AD relative to nondemented control brain.17 Taken together, these data imply an overall negative role for excessive TNF signaling on AD pathophysiology but, perhaps more importantly, illustrate the complexity of this signaling pathway.Despite a large body of literature indicating detrimental roles for TNF-α, neuroprotective effects have also been reported. Early experiments revealed that TNF-α is protective in cultured neurons during glucose deprivation–induced injury and excitotoxicity by preserving Ca2+ homeostasis.18 Barger et al19 further demonstrated in dissociated neuronal cultures that pretreatment with TNF-α and Aβ peptide spares cells from Aβ-induced neuronal death, iron toxicity, and intracellular Ca2+ accumulation via an NF-κB–dependent mechanism. Moreover, neurons are vulnerable to ischemic injury and oxidative stress in TNF-R null mice, indicating that TNF-α is protective.20 Mice lacking TNF-R expression exhibited reduced manganese superoxide dismutase activity and lacked a robust microglial response to kainic acid.20 Similarly, cultured neurons pretreated with TNF-α resulted in a significant increase in manganese superoxide dismutase activity and a reduction in superoxide accumulation.21 These data add to the complexity of the TNF signaling pathway and suggest that strategies to modulate TNF-α in the disease setting may require selective tuning and specificity to ensure that protective signaling outcomes are not compromised.Nonetheless, given the compelling data supporting the pathologic role of TNF-α in AD, the potential of using anti–TNF-α therapeutics has become a viable strategy for subverting the disease course. Preclinical data by McAlpine et al22 demonstrate that transiently inhibiting soluble TNF signaling in the 3xTg-AD mouse model using a dominant-negative inhibitor in conjunction with enhanced systemic inflammation prevents AD-associated amyloid pathology. Tobinick et al23 reported in a short-term, prospective, open-label pilot study that semiweekly perispinal administration of etanercept, a receptor decoy biological agent antagonizing the actions of TNF-α, in 15 patients with mild to severe AD led to significant and rapid cognitive improvements compared with untreated control patients as assessed by three separate tests measuring cognitive function.Although previous studies provide evidence suggesting that TNF-α inhibition in the short-term may lead to improved pathologic and functional outcomes, they lack data addressing the long-term consequences of blocking TNF-α in a global manner, where cell, stage, and receptor specificity were not examined. To this end, we recently demonstrated that long-term global inhibition of TNF-R signaling in 3xTg-AD mice where TNF-RI and TNF-RII were ablated in all cell types results in a robust increase in hallmark amyloid and NFT pathology. Furthermore, in the absence of TNF signaling, microglia seem nonresponsive to the developing amyloid pathology, which correlates with an impairment of microglial-mediated Aβ42 phagocytosis activity in vitro.24 These data suggest that caution should be taken with the use of broad long-term anti-TNF inhibitors and that a more selective strategy should be investigated.To add to our understanding of TNF signaling biology and the consequences of selectively modulating this pathway, we investigated the cell- and stage-specific role of TNF-R signaling in AD by using recombinant AAV (rAAV) vector–delivered siRNA technology to selectively knock down neuronal TNF-R signaling at stages preceding progressive pathology or in the presence of extant disease using the 3xTg-AD mouse model. We demonstrate that neuronal TNF-RI and TNF-RII exert differential actions where intact TNF-RII signaling results in suppressed Aβ plaque deposition and paired helical filament (PHF) formation in the context of progressive and established disease pathogenesis. In addition, we report a substantial reduction in Iba-1–positive microglia when rAAV2-delivered siTNF-RII or siTNF-RI+RII viral vectors are administered at 2 and 12 months of age. Taken together, these data demonstrate that selectively suppressing neuronal TNF-RI and/or TNF-RII leads to distinct and significant changes in AD pathogenesis, which is most likely a consequence of the divergent signaling pathways associated with these receptors. The present findings support further development and rigorous study of highly selective strategies designed to inhibit specific TNF-α–mediated signals and potentially disrupt the onset and/or progression of this debilitating disease.  相似文献   
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号