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   10378篇
  免费   955篇
  国内免费   437篇
耳鼻咽喉   126篇
儿科学   115篇
妇产科学   224篇
基础医学   2017篇
口腔科学   314篇
临床医学   719篇
内科学   1284篇
皮肤病学   318篇
神经病学   226篇
特种医学   198篇
外国民族医学   1篇
外科学   783篇
综合类   1877篇
现状与发展   1篇
预防医学   708篇
眼科学   84篇
药学   803篇
  5篇
中国医学   285篇
肿瘤学   1682篇
  2023年   112篇
  2022年   199篇
  2021年   307篇
  2020年   326篇
  2019年   361篇
  2018年   308篇
  2017年   330篇
  2016年   314篇
  2015年   366篇
  2014年   567篇
  2013年   558篇
  2012年   528篇
  2011年   714篇
  2010年   561篇
  2009年   555篇
  2008年   584篇
  2007年   635篇
  2006年   588篇
  2005年   456篇
  2004年   453篇
  2003年   423篇
  2002年   372篇
  2001年   374篇
  2000年   379篇
  1999年   290篇
  1998年   167篇
  1997年   133篇
  1996年   95篇
  1995年   83篇
  1994年   68篇
  1993年   54篇
  1992年   62篇
  1991年   30篇
  1990年   32篇
  1989年   29篇
  1988年   28篇
  1987年   23篇
  1986年   16篇
  1985年   37篇
  1984年   41篇
  1983年   20篇
  1982年   37篇
  1981年   29篇
  1980年   22篇
  1979年   17篇
  1978年   18篇
  1977年   9篇
  1976年   12篇
  1975年   13篇
  1974年   11篇
排序方式: 共有10000条查询结果,搜索用时 15 毫秒
91.
目的:观察电针防治干预对动脉粥样硬化(AS)动脉斑块及科水平肠道菌群结构特征的影响,筛选特征性菌群,探析电针抗AS的机制。方法:实验兔平均随机分为空白组、模型组、防治组,每组6只。模型组以高脂饲料结合颈总动脉球囊损伤术方法造模;防治组先予电针干预后造模,后再予电针干预。观察油红O染色、16S rRNA测序与生信分析结果。结果:防治组颈动脉内壁沉积脂质斑块明显少于模型组;空白组与防治组组间肠道菌群群落差异更小;防治组S24-7、Synergistaceae、Veillonellaceae、[Mogibacteriaceae]科菌群相对丰度与模型组比较,差异有统计学意义(P<0.05,P<0.01),与空白组相近。结论:AS与肠道菌群结构变化有关,电针能抑制动脉斑块形成并调整肠道菌群结构,可能与S24-7 Veillonellaceae、[Mogibacteriaceae]科菌群相对丰度有关。  相似文献   
92.
目的:探究不同月龄APP/PS1双转基因模型小鼠的肠道菌群特点及补肾法对其肠道菌群失衡的影响。方法:将8只6个月龄雄性APP/PS1小鼠随机分为模型组和补肾组;4只6个月龄雄性C57BL/6小鼠作为正常对照组。补肾组小鼠给予补肾法中药灌胃3个月,模型组和正常对照组小鼠均予以等体积羧甲基纤维素(CMC)溶液灌胃。留取小鼠灌胃干预前后的粪便,运用16SrDNA技术检测各组肠道菌群的组成结构。从肠道微生态方面,探讨补肾中药对APP/PS1双转基因小鼠肠道菌群构成的影响。结果:6个月龄时模型组和正常对照组小鼠的肠道菌群差异无统计学意义;与9个月龄正常组比较,模型组小鼠肠道内芽孢杆菌纲丰度明显升高。与模型组比较,补肾组肠道内芽孢杆菌纲丰度明显下降,疣微菌门丰度呈上升趋势。结论:APP/PS1小鼠体内均存在肠道菌群失衡的情况,主要与芽孢菌纲、疣微菌门的丰度水平的变化相关。补肾法可以通过降低芽孢菌纲、升高疣微菌门的丰度来进一步改善AD小鼠肠道菌群失衡状态。  相似文献   
93.
We develop a rapid (<4 h) and reliable multiplex polymerase chain reaction for screening of 16S rRNA methylase genes conferring resistance to aminoglycosides. This study particularly underlined that 16S rRNA methylases are frequently (75%) identified among Enterobacteriaceae isolates producing the carbapenemase NDM-1.  相似文献   
94.
95.

Introduction

The association of thrombotic thrombocytopenic purpura (TTP) with systemic lupus erythematosus (SLE) is rare. It is associated with high morbidity and mortality. Information about risk factors and clinical outcomes is scant.

Material and Methods

A retrospective case-control study was performed in a referral center in Mexico City between 1994 and 2013. Patients were diagnosed with TTP if they fulfilled the following criteria: microangiopathic haemolytic anaemia, thrombocytopenia, high LDH levels, normal fibrinogen and negative Coombs’ test. Patients with SLE were diagnosed with ≥ 4 ACR criteria. We included three study groups: group A included patients with SLE-associated TTP (TTP/SLE; cases n = 22, TTP events n = 24); patients with non-autoimmune TTP (NA-TTP; cases n = 19, TTP events n = 22) were included in group B and patients with SLE without TTP (n = 48) in group C.

Results

After multivariate analysis, lymphopenia < 1000/mm3 [OR 19.84, p = 0.037], high SLEDAI score three months prior to hospitalisation [OR 1.54, p = 0.028], Hg < 7 g/dL [OR 6.81, p = 0.026], low levels of indirect bilirubin [OR 0.51, p = 0.007], and less severe thrombocytopenia [OR 0.98, p = 0.009] were associated with TTP in SLE patients. Patients with TTP/SLE received increased cumulative steroid dose vs. NA-TTP (p = 0.006) and a higher number of immunosuppressive drugs (p = 0.015). Patients with TTP/SLE had higher survival than NA-TTP (p = 0.033); however, patients hospitalised for TTP/SLE had a higher risk of death than lupus patients hospitalised for other causes

Conclusions

Lymphopenia is an independent risk factor for TTP/SLE. It is likely that patients with TTP/SLE present with less evident clinical features, so the level of suspicion must be higher to avoid delay in treatment.  相似文献   
96.
目的:探讨p16、p53、Cerb B2和ki-67在慢性胃炎、异型增生、胃癌及癌旁组织中的表达及临床意义。方法:采用免疫组织化学SP法对慢性胃炎组织、异型增生组织、胃癌组织及癌旁组织分别行p16、p53、Cerb B2和ki-67检测。结果:p16在慢性胃炎、低级别异型增生、高级别异型增生、胃癌及癌旁组织中的表达阳性率分别为25.49%、30.39%、33.93%、33.55%和32.69%。p53在慢性胃炎、低级别异型增生、高级别异型增生、胃癌及癌旁组织中的表达阳性率分别为0、0、21.43%、50.00%和13.46%。Cerb B-2在慢性胃炎、低级别异型增生、高级别异型增生、胃癌及癌旁组织中的表达阳性率分别为0、0、14.29%、25.66%和9.62%。ki-67在慢性胃炎、低级别异型增生、高级别异型增生、胃癌及癌旁组织中的表达阳性率分别为15.69%、43.14%、57.14%、75.66%和42.31%。结论:p16、p53、Cerb B-2和ki-67在癌前病变中异常表达,可作为判断胃癌及癌前病变的重要参考指标。  相似文献   
97.
98.
99.
100.
Ribosomopathies are a class of diseases caused by mutations that affect the biosynthesis and/or functionality of the ribosome. Although they initially present as hypoproliferative disorders, such as anemia, patients have elevated risk of hyperproliferative disease (cancer) by midlife. Here, this paradox is explored using the rpL10-R98S (uL16-R98S) mutant yeast model of the most commonly identified ribosomal mutation in acute lymphoblastic T-cell leukemia. This mutation causes a late-stage 60S subunit maturation failure that targets mutant ribosomes for degradation. The resulting deficit in ribosomes causes the hypoproliferative phenotype. This 60S subunit shortage, in turn, exerts pressure on cells to select for suppressors of the ribosome biogenesis defect, allowing them to reestablish normal levels of ribosome production and cell proliferation. However, suppression at this step releases structurally and functionally defective ribosomes into the translationally active pool, and the translational fidelity defects of these mutants culminate in destabilization of selected mRNAs and shortened telomeres. We suggest that in exchange for resolving their short-term ribosome deficits through compensatory trans-acting suppressors, cells are penalized in the long term by changes in gene expression that ultimately undermine cellular homeostasis.Ribosomopathies are a family of congenital diseases that are linked to genetic defects in ribosomal proteins or ribosome biogenesis factors. They are characterized by pleiotropic abnormalities that include birth defects, heart and lung diseases, connective tissue disorders, anemia, ataxia, and mental retardation (reviewed in ref. 1). Although each ribosomopathy presents a unique pathological spectrum, the inherited forms are characterized by bone marrow failure and anemia early in life, followed by elevated cancer risk by middle age. For example, although childhood anemia is one of the cardinal symptoms of the genetically inherited disease Diamond–Blackfan anemia, these patients have a fivefold higher lifetime risk of cancer than the general population and a 30- to 40-fold higher risk of developing acute myeloid leukemia, osteosarcoma, or colon cancer (reviewed in refs. 2, 3). Similarly, patients with X-linked dyskeratosis are predisposed to myeloid leukemia and a variety of solid tumors (4), whereas patients with 5q− syndrome are at higher risk of developing acute myeloid leukemia (reviewed in ref. 5). In the genetically tractable zebrafish model, heterozygous loss-of-function mutations in several ribosomal proteins cause development of peripheral nerve sheet tumors (6). Somatically acquired mutations in ribosomal proteins are also implicated in cancer: ∼10% of children with T-cell acute lymphoblastic leukemia (T-ALL) were found to harbor somatic mutations in the ribosomal protein of the large subunit (LSU) 10, 5, and 22 (RPL10, RPL5, and RPL22) (7). [Note that the proteins encoded by these genes are also named uL16, uL18, and eL22, respectively, under the newly proposed uniform ribosomal protein nomenclature (8).] A separate study identified heterozygous deletions in the region of chromosome 1p that contains RPL22 (eL22) in an additional 10% of patients with T-ALL (9). The model of ribosomal proteins as targets for somatic mutations in cancer is further supported by the finding that two ribosomal protein genes (RPL5/uL18 and RPL22/eL22) are included in the list of 127 genes identified as significantly mutated in cancer in the context of the first Cancer Genome Atlas pan-cancer analysis in 12 tumor types (10).Ribosomopathies present an intriguing paradox: Although patients initially present with hypoproliferative disorders (e.g., anemias, bone marrow failure), those who survive to middle age often develop hyperproliferative diseases (i.e., cancers). The link between ribosome defects and hypoproliferative disease phenotypes has been extensively studied: The current working hypothesis is that impaired ribosome biogenesis activates a “ribosomal stress” cascade, activating the cellular TP53 pathway and resulting in cell cycle arrest and cell death (11). However, activation of TP53 does not explain why ribosomal defects are associated with hyperproliferative diseases, particularly cancer. Mutations in the ribosomal protein gene RPL10/uL16 were recently identified in patients with T-ALL (7). The T-ALL–associated RPL10/uL16 mutations occurred almost exclusively in residue arginine 98 (R98), with the exception of one patient harboring the Q123P mutation, which lies adjacent to R98 within the rpL10/uL16 3D structure (Fig. 1). Both residues are at the base of an essential flexible loop in rpL10 that closely approaches the peptidyltransferase center in the catalytic core in the ribosome (12). In addition to its role in catalysis (13, 14), rpL10/uL16 plays an important role in the late stages of 60S subunit biogenesis. After initial production of the separate ribosomal subunits in the nucleus, immature and functionally inactive pre-60S subunits are exported to the cytoplasm, where they undergo additional maturation events (15), including incorporation of rpL10/uL16, before they can associate with mature 40S subunits and engage in protein synthesis (16). Among the critical set of final 60S maturation steps is the release of the antiassociation factor Tif6, followed by release of Nmd3, the primary export adaptor for the pre-60S subunit in yeast and in humans (17, 18). Tif6 release requires the tRNA structural mimic Sdo1p (19) and the GTPase Efl1, a paralog of eukaryotic elongation factor 2 (eEF2) (20). We have suggested that structural rearrangements of the internal loop of rpL10/uL16 coordinate this final maturation process, resulting in a test drive of the pre-60S subunit to ensure that only properly functioning subunits are allowed to enter the pool of translationally active ribosomes (13, 21). Defective ribosomes carrying mutations in rpL10/uL16 specifically fail in this test drive, leading to their degradation through a molecular pathway that is yet to be characterized. Beyond 60S maturation, rpL10/uL16 plays an important role in coordinating intersubunit rotation and controlling allosteric rearrangements within the ribosome, helping to ensure the directionality and fidelity of protein synthesis (13).Open in a separate windowFig. 1.Localization of rpL10 and the loop in the LSU. (A) rpL10/uL16 in the context of the crown view of the LSU. (B) Close-up view of rpL10/uL16 and the local environment. The flexible loop structure is indicated by dashed red lines, and the positions of R98 and Q123 are indicated. rpL10/uL16 is situated between helices 38 and 89, and it is located in close proximity to several functional centers of the LSU, including the peptidyltransferase center (PTC), aa-tRNA accommodation corridor, and elongation factor binding site. Images were generated using PyMOL.rpL10/uL16 is highly conserved among eukaryotes: The yeast and human proteins are interchangeable, and residue 98 is invariantly an arginine (22). Human RPL10/uL16 is located on the X chromosome, and is therefore expressed as a single-copy gene in males. Thus, the haploid yeast model is an excellent mimic of the situation in the cells of a patient with T-ALL. Yeast cells expressing rpl10-R98S, rpl10-R98C, and rpl10-H123P (corresponding to Q123 in human rpL10/uL16) as the sole forms of rpL10/uL16 displayed proliferative defects. Further, polysome profiling revealed increased ratios of free 60S and 40S subunits vs. monosomes, markedly reduced polysomes, and the presence of halfmers in these mutants, suggesting defects in both ribosome biogenesis and subunit joining (7). Tif6 and Nmd3 both accumulated in the cytoplasm in the mutant cells, indicating a defect in their release from the cytoplasmic 60S (7). Thus, all of the rpl10/uL16 mutations appeared to affect 60S biogenesis at the Efl1-dependent quality control step. Consistent with the yeast-based observations, mouse lymphoid cells expressing rpl10-R98S displayed slower proliferation rates than cells expressing WT RPL10/uL16 and conferred defective polysome profiles (7).The studies presented in the current report use the yeast rpl10-R98S mutant to elucidate the structural, biochemical, and trans-lational fidelity defects that may lead to carcinogenesis. This mutant perturbs the structural equilibrium of ribosomes toward the “rotated state.” At the biochemical level, this underlying structural defect alters the affinity of mutant ribosomes for a specific set of trans-acting ligands. In turn, the biochemical defects affect translational fidelity, promoting elevated rates of −1 programmed ribosomal frameshifting (−1 PRF) and impaired recognition of termination codons. Globally increased rates of −1 PRF result in a decreased abundance of cellular mRNAs that harbor operational −1 PRF signals (23, 24). These −1 PRF signal-containing mRNAs include EST1, EST2, STN1, and CDC13, which play central roles in yeast telomere maintenance (23). In rpl10-R98S cells, the steady-state abundances of these mRNAs are decreased, resulting in telomere shortening. A spontaneously acquired trans-acting mutant suppresses the ribosome biogenesis defects of the rpl10-R98S mutant, thereby reestablishing high levels of ribosome production and cell proliferation. Importantly, however, suppression of the biogenesis and growth impairment defects fails to suppress the profound structural, biochemical, and translational fidelity defects of rpL10-R98S ribosomes. These findings suggest that suppression of the growth defect results from bypassing the test drive. Although the suppressor mutation enables cells to grow at normal rates, genetic suppression comes at the cost of releasing functionally defective ribosomes into the translationally active pool. We propose two different but not mutually exclusive models for how somatically acquired rpL10/uL16 mutations may promote cancer: (i) Mutant ribosomes may drive altered gene expression programs, promoting T-ALL, or (ii) the suppressor mutations may themselves be the drivers of T-ALL.  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