细胞程序性死亡的机制

IF 7.5 2区 医学 Q1 IMMUNOLOGY Immunological Reviews Pub Date : 2023-12-14 DOI:10.1111/imr.13303
Tian Li, Guido Kroemer
{"title":"细胞程序性死亡的机制","authors":"Tian Li,&nbsp;Guido Kroemer","doi":"10.1111/imr.13303","DOIUrl":null,"url":null,"abstract":"<p>The present volume of <i>Immunological Reviews</i> deals with the mechanisms of programmed cell death, obviously from an immunological perspective. What are the consequences of cell death on the organism and, in particular, on the immune recognition of stressed and dying cells? The long-distance effects of therapeutic manipulations resulting in the death of cancer cells are surprisingly vast, as this has been documented for the treatment with clinically approved or experimental chemotherapeutic agents. For example, cell death can cause neuropathic pain through mechanisms of neuroinflammation.<span><sup>1</sup></span> In addition, cell death induction can result in the production of Type I interferons by tumor cells that then mediate ambiguous adaptive responses ranging from an enhancement of cancer cell stemness and exhaustion of anticancer immune response within the tumor microenvironment to the stimulation of anticancer immune responses. Type I interferon can even trigger a systemic sickness response ranging from flu-like symptoms to a state of depression.<span><sup>2</sup></span> Such long-range effects of cell death are certainly also relevant to the pathophysiology of viral infections.</p><p>If induced in an appropriate fashion, one of the major positive effects of cancer cell stress and death is the induction of immune responses against tumor-associated antigen, thus sensitizing tumors to immunotherapy with immune checkpoint inhibitors.<span><sup>3-5</sup></span> This has important therapeutic implications because chemotherapeutics that induce immunogenic cell death can be used as first-line treatments to sensitize major cancer types (exemplified by KRAS-mutated colorectal cancer, non-small cell lung cancer and triple-negative breast cancer) to subsequent immunotherapy with antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or PD-1 ligand-1 (PD-L1), as this has been confirmed in several clinical trials.</p><p>Of note, there are multiple different subroutines of cell death, and several if not all of them can be immunogenic, as this has been documented for apoptosis (which involves mitochondrial membrane permeabilization and the activation of caspases 3 and 7)<span><sup>2, 3</sup></span> but also for necroptosis (with the implication of specific effector molecules including receptor-interacting kinase 3 (RIP3) and mixed lineage kinase domain-like pseudokinase (MLKL1)),<span><sup>6</sup></span> pyroptosis (involving inflammasome/caspase-1-mediated activation of pore-forming gasdermins),<span><sup>7</sup></span> a mixture of pyroptosis, apoptosis, and necroptosis dubbed PANoptosis,<span><sup>8</sup></span> ferroptosis (involving lethal membrane damage by peroxidation),<span><sup>9, 10</sup></span> and cuproptosis (due to copper-induced aggregation of lipoylated dihydrolipoamide S-acetyltransferase).<span><sup>11</sup></span> In all cases, cell death can be preceded by immunogenic stress that favors the emission of danger-associated molecular patterns (DAMPs) appearing on the surface of the cells or secreted into the extracellular space. It is the sum of stress-associated DAMPs (that are surface-exposed or released before cells disintegrate) and that of death-associated DAMPs (that become accessible or are passively released when the plasma membrane and internal membrane of cells become permeable) that dictates the immunogenicity of cell death and hence the capacity of the immune system to detect dead cell antigens. Such antigens can be microbial (for instance in the context of infection by viruses or intracellular bacteria), tumor-associated, or autoantigens.</p><p>Immunogenic cell death is not only induced by drugs but can also occur in the context of radiation therapy,<span><sup>12</sup></span> photodynamic, and photothermal therapy,<span><sup>13</sup></span> as well upon infection by microbes including oncolytic viruses.<span><sup>14</sup></span> Logically, attempts are underway to create novel galenic formulation including nanoparticle-based drug delivery systems to administer drugs that induce immunogenic cell death in tumors, yet do not mediate any systemic effects.<span><sup>13, 15</sup></span> Interestingly, cell death of cancer cells can be accompanied by the release of nanoscale extracellular vesicles dubbed exosomes that constitute potential biomarkers of ongoing cell death events and establish short- and long-distance communication with neighboring cells and distant tissues.<span><sup>16</sup></span> As a possibility, such exosomes might be engineered for the nanodelivery of therapeutic agents.</p><p>When cells undergo immunogenic stress and death, they interact primarily with dendritic cells,<span><sup>17</sup></span> in particular with Type-1 conventional dendritic cells (cDC1) that appear to be particularly competent in eliciting responses against dead cell antigens.<span><sup>18</sup></span> Dendritic cells can be loaded with stressed and dying cancer cells and then be used as prophylactic or therapeutic vaccines.<span><sup>17</sup></span> Moreover, dendritic cells can be manipulated pharmacologically to enhance their capacity to present tumor antigens to T cells.<span><sup>18</sup></span> Such dendritic cells educate cytotoxic T lymphocytes to recognize and lyse malignant cells. Importantly, this process of T-cell-mediated cytotoxicity can elicit immunogenic cell death, hence amplifying the phenomenon and protracting the anticancer immune response.<span><sup>19</sup></span> However, cell death affecting immune cells may play down such a desirable immunosurveillance. Specifically, it appears that dying neutrophil granulocytes produce so-called neutrophil extracellular traps (NETs) that shield cancer cells from cytotoxic immunity, hence impairing their clearance.<span><sup>20</sup></span> Moreover, dying neutrophils can stimulate unwarranted inflammatory and autoimmune responses.<span><sup>21</sup></span></p><p>Altogether, this volume of <i>Immunological Reviews</i> illustrates to which extent different cell stress and death modalities affecting malignant cells, pathogen-infected cells, or immune cells can elicit innate and cognate immune responses with vast consequences for whole-body physiology. It appears that processes that for long have been studied exclusively by cell biologists have acquired a major immunological dimension that already yields tangible impact with respect to the clinical management of malignant diseases. Future will tell whether the knowledge generated in this field will also contribute to the prevention and treatment of infectious and autoimmune diseases.</p><p>The authors declare no relevant conflicts of interest.</p>","PeriodicalId":178,"journal":{"name":"Immunological Reviews","volume":null,"pages":null},"PeriodicalIF":7.5000,"publicationDate":"2023-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imr.13303","citationCount":"0","resultStr":"{\"title\":\"Mechanisms of programmed cell death\",\"authors\":\"Tian Li,&nbsp;Guido Kroemer\",\"doi\":\"10.1111/imr.13303\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>The present volume of <i>Immunological Reviews</i> deals with the mechanisms of programmed cell death, obviously from an immunological perspective. What are the consequences of cell death on the organism and, in particular, on the immune recognition of stressed and dying cells? The long-distance effects of therapeutic manipulations resulting in the death of cancer cells are surprisingly vast, as this has been documented for the treatment with clinically approved or experimental chemotherapeutic agents. For example, cell death can cause neuropathic pain through mechanisms of neuroinflammation.<span><sup>1</sup></span> In addition, cell death induction can result in the production of Type I interferons by tumor cells that then mediate ambiguous adaptive responses ranging from an enhancement of cancer cell stemness and exhaustion of anticancer immune response within the tumor microenvironment to the stimulation of anticancer immune responses. Type I interferon can even trigger a systemic sickness response ranging from flu-like symptoms to a state of depression.<span><sup>2</sup></span> Such long-range effects of cell death are certainly also relevant to the pathophysiology of viral infections.</p><p>If induced in an appropriate fashion, one of the major positive effects of cancer cell stress and death is the induction of immune responses against tumor-associated antigen, thus sensitizing tumors to immunotherapy with immune checkpoint inhibitors.<span><sup>3-5</sup></span> This has important therapeutic implications because chemotherapeutics that induce immunogenic cell death can be used as first-line treatments to sensitize major cancer types (exemplified by KRAS-mutated colorectal cancer, non-small cell lung cancer and triple-negative breast cancer) to subsequent immunotherapy with antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or PD-1 ligand-1 (PD-L1), as this has been confirmed in several clinical trials.</p><p>Of note, there are multiple different subroutines of cell death, and several if not all of them can be immunogenic, as this has been documented for apoptosis (which involves mitochondrial membrane permeabilization and the activation of caspases 3 and 7)<span><sup>2, 3</sup></span> but also for necroptosis (with the implication of specific effector molecules including receptor-interacting kinase 3 (RIP3) and mixed lineage kinase domain-like pseudokinase (MLKL1)),<span><sup>6</sup></span> pyroptosis (involving inflammasome/caspase-1-mediated activation of pore-forming gasdermins),<span><sup>7</sup></span> a mixture of pyroptosis, apoptosis, and necroptosis dubbed PANoptosis,<span><sup>8</sup></span> ferroptosis (involving lethal membrane damage by peroxidation),<span><sup>9, 10</sup></span> and cuproptosis (due to copper-induced aggregation of lipoylated dihydrolipoamide S-acetyltransferase).<span><sup>11</sup></span> In all cases, cell death can be preceded by immunogenic stress that favors the emission of danger-associated molecular patterns (DAMPs) appearing on the surface of the cells or secreted into the extracellular space. It is the sum of stress-associated DAMPs (that are surface-exposed or released before cells disintegrate) and that of death-associated DAMPs (that become accessible or are passively released when the plasma membrane and internal membrane of cells become permeable) that dictates the immunogenicity of cell death and hence the capacity of the immune system to detect dead cell antigens. Such antigens can be microbial (for instance in the context of infection by viruses or intracellular bacteria), tumor-associated, or autoantigens.</p><p>Immunogenic cell death is not only induced by drugs but can also occur in the context of radiation therapy,<span><sup>12</sup></span> photodynamic, and photothermal therapy,<span><sup>13</sup></span> as well upon infection by microbes including oncolytic viruses.<span><sup>14</sup></span> Logically, attempts are underway to create novel galenic formulation including nanoparticle-based drug delivery systems to administer drugs that induce immunogenic cell death in tumors, yet do not mediate any systemic effects.<span><sup>13, 15</sup></span> Interestingly, cell death of cancer cells can be accompanied by the release of nanoscale extracellular vesicles dubbed exosomes that constitute potential biomarkers of ongoing cell death events and establish short- and long-distance communication with neighboring cells and distant tissues.<span><sup>16</sup></span> As a possibility, such exosomes might be engineered for the nanodelivery of therapeutic agents.</p><p>When cells undergo immunogenic stress and death, they interact primarily with dendritic cells,<span><sup>17</sup></span> in particular with Type-1 conventional dendritic cells (cDC1) that appear to be particularly competent in eliciting responses against dead cell antigens.<span><sup>18</sup></span> Dendritic cells can be loaded with stressed and dying cancer cells and then be used as prophylactic or therapeutic vaccines.<span><sup>17</sup></span> Moreover, dendritic cells can be manipulated pharmacologically to enhance their capacity to present tumor antigens to T cells.<span><sup>18</sup></span> Such dendritic cells educate cytotoxic T lymphocytes to recognize and lyse malignant cells. Importantly, this process of T-cell-mediated cytotoxicity can elicit immunogenic cell death, hence amplifying the phenomenon and protracting the anticancer immune response.<span><sup>19</sup></span> However, cell death affecting immune cells may play down such a desirable immunosurveillance. Specifically, it appears that dying neutrophil granulocytes produce so-called neutrophil extracellular traps (NETs) that shield cancer cells from cytotoxic immunity, hence impairing their clearance.<span><sup>20</sup></span> Moreover, dying neutrophils can stimulate unwarranted inflammatory and autoimmune responses.<span><sup>21</sup></span></p><p>Altogether, this volume of <i>Immunological Reviews</i> illustrates to which extent different cell stress and death modalities affecting malignant cells, pathogen-infected cells, or immune cells can elicit innate and cognate immune responses with vast consequences for whole-body physiology. It appears that processes that for long have been studied exclusively by cell biologists have acquired a major immunological dimension that already yields tangible impact with respect to the clinical management of malignant diseases. Future will tell whether the knowledge generated in this field will also contribute to the prevention and treatment of infectious and autoimmune diseases.</p><p>The authors declare no relevant conflicts of interest.</p>\",\"PeriodicalId\":178,\"journal\":{\"name\":\"Immunological Reviews\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":7.5000,\"publicationDate\":\"2023-12-14\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imr.13303\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Immunological Reviews\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1111/imr.13303\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Immunological Reviews","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1111/imr.13303","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

本卷免疫学评论处理程序性细胞死亡的机制,显然是从免疫学的角度。细胞死亡对机体,特别是对应激细胞和死亡细胞的免疫识别有什么影响?治疗操作导致癌细胞死亡的远距离影响是惊人的巨大,因为这已经被临床批准或实验化疗药物的治疗所记录。例如,细胞死亡可通过神经炎症机制引起神经性疼痛此外,细胞死亡诱导可导致肿瘤细胞产生I型干扰素,然后介导模棱两可的适应性反应,从肿瘤微环境中癌细胞干细胞性的增强和抗癌免疫反应的耗尽到抗癌免疫反应的刺激。I型干扰素甚至可以引发从流感样症状到抑郁状态的全身性疾病反应细胞死亡的这种长期影响当然也与病毒感染的病理生理学有关。如果以适当的方式诱导,癌细胞应激和死亡的主要积极作用之一是诱导针对肿瘤相关抗原的免疫反应,从而使肿瘤对免疫检查点抑制剂的免疫治疗敏感。3-5这具有重要的治疗意义,因为诱导免疫原性细胞死亡的化疗药物可以作为一线治疗方法,使主要癌症类型(例如kras突变的结直肠癌、非小细胞肺癌和三阴性乳腺癌)对随后使用靶向细胞毒性t淋巴细胞相关蛋白4 (CTLA-4)、程序性细胞死亡1 (PD-1)或PD-1配体-1 (PD-L1)的抗体进行免疫治疗变得敏感。因为这已经在几个临床试验中得到了证实。值得注意的是,细胞死亡有多种不同的子程序,其中一些(如果不是全部的话)可以是免疫原性的,因为这已经被证明是细胞凋亡(涉及线粒体膜渗透和半胱天冬酶3和7的激活)2。也可用于坏死性死亡(包括受体相互作用激酶3 (RIP3)和混合谱系激酶结构域样伪激酶(MLKL1)等特定效应分子),6焦亡(涉及炎症小体/caspase-1介导的成孔gasdermins的激活),7焦亡,细胞凋亡和坏死性死亡的混合物,称为PANoptosis,8铁亡(涉及过氧化致死膜损伤),9。10和铜还原(由于铜诱导的脂化二氢脂酰胺s -乙酰转移酶聚集)在所有情况下,细胞死亡之前都可能发生免疫原性应激,这种应激有利于释放出现在细胞表面或分泌到细胞外空间的危险相关分子模式(DAMPs)。应激相关的DAMPs(在细胞解体前表面暴露或释放)和死亡相关的DAMPs(当细胞的质膜和细胞膜变得可透性时变得可接近或被动释放)的总和决定了细胞死亡的免疫原性,从而决定了免疫系统检测死亡细胞抗原的能力。这些抗原可以是微生物抗原(例如在病毒或细胞内细菌感染的情况下)、肿瘤相关抗原或自身抗原。免疫原性细胞死亡不仅可由药物引起,也可在放射治疗、光动力疗法和光热疗法13以及溶瘤病毒等微生物感染的情况下发生从逻辑上讲,人们正在尝试创造新的galenic配方,包括基于纳米颗粒的药物递送系统,以给药,诱导肿瘤中免疫原性细胞死亡,但不介导任何全身效应。有趣的是,癌细胞的死亡可能伴随着被称为外泌体的纳米级细胞外囊泡的释放,这些外泌体构成正在进行的细胞死亡事件的潜在生物标志物,并与邻近细胞和远处组织建立短距离和远距离通信作为一种可能性,这种外泌体可能被设计用于治疗药物的纳米递送。当细胞遭受免疫原性应激和死亡时,它们主要与树突状细胞相互作用17,特别是与1型常规树突状细胞(cDC1)相互作用,后者似乎特别有能力引发针对死细胞抗原的反应18树突状细胞可以装载压力和垂死的癌细胞,然后用作预防性或治疗性疫苗此外,树突状细胞可以通过药理学手段增强其向T细胞呈递肿瘤抗原的能力这种树突状细胞诱导细胞毒性T淋巴细胞识别并溶解恶性细胞。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Mechanisms of programmed cell death

The present volume of Immunological Reviews deals with the mechanisms of programmed cell death, obviously from an immunological perspective. What are the consequences of cell death on the organism and, in particular, on the immune recognition of stressed and dying cells? The long-distance effects of therapeutic manipulations resulting in the death of cancer cells are surprisingly vast, as this has been documented for the treatment with clinically approved or experimental chemotherapeutic agents. For example, cell death can cause neuropathic pain through mechanisms of neuroinflammation.1 In addition, cell death induction can result in the production of Type I interferons by tumor cells that then mediate ambiguous adaptive responses ranging from an enhancement of cancer cell stemness and exhaustion of anticancer immune response within the tumor microenvironment to the stimulation of anticancer immune responses. Type I interferon can even trigger a systemic sickness response ranging from flu-like symptoms to a state of depression.2 Such long-range effects of cell death are certainly also relevant to the pathophysiology of viral infections.

If induced in an appropriate fashion, one of the major positive effects of cancer cell stress and death is the induction of immune responses against tumor-associated antigen, thus sensitizing tumors to immunotherapy with immune checkpoint inhibitors.3-5 This has important therapeutic implications because chemotherapeutics that induce immunogenic cell death can be used as first-line treatments to sensitize major cancer types (exemplified by KRAS-mutated colorectal cancer, non-small cell lung cancer and triple-negative breast cancer) to subsequent immunotherapy with antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or PD-1 ligand-1 (PD-L1), as this has been confirmed in several clinical trials.

Of note, there are multiple different subroutines of cell death, and several if not all of them can be immunogenic, as this has been documented for apoptosis (which involves mitochondrial membrane permeabilization and the activation of caspases 3 and 7)2, 3 but also for necroptosis (with the implication of specific effector molecules including receptor-interacting kinase 3 (RIP3) and mixed lineage kinase domain-like pseudokinase (MLKL1)),6 pyroptosis (involving inflammasome/caspase-1-mediated activation of pore-forming gasdermins),7 a mixture of pyroptosis, apoptosis, and necroptosis dubbed PANoptosis,8 ferroptosis (involving lethal membrane damage by peroxidation),9, 10 and cuproptosis (due to copper-induced aggregation of lipoylated dihydrolipoamide S-acetyltransferase).11 In all cases, cell death can be preceded by immunogenic stress that favors the emission of danger-associated molecular patterns (DAMPs) appearing on the surface of the cells or secreted into the extracellular space. It is the sum of stress-associated DAMPs (that are surface-exposed or released before cells disintegrate) and that of death-associated DAMPs (that become accessible or are passively released when the plasma membrane and internal membrane of cells become permeable) that dictates the immunogenicity of cell death and hence the capacity of the immune system to detect dead cell antigens. Such antigens can be microbial (for instance in the context of infection by viruses or intracellular bacteria), tumor-associated, or autoantigens.

Immunogenic cell death is not only induced by drugs but can also occur in the context of radiation therapy,12 photodynamic, and photothermal therapy,13 as well upon infection by microbes including oncolytic viruses.14 Logically, attempts are underway to create novel galenic formulation including nanoparticle-based drug delivery systems to administer drugs that induce immunogenic cell death in tumors, yet do not mediate any systemic effects.13, 15 Interestingly, cell death of cancer cells can be accompanied by the release of nanoscale extracellular vesicles dubbed exosomes that constitute potential biomarkers of ongoing cell death events and establish short- and long-distance communication with neighboring cells and distant tissues.16 As a possibility, such exosomes might be engineered for the nanodelivery of therapeutic agents.

When cells undergo immunogenic stress and death, they interact primarily with dendritic cells,17 in particular with Type-1 conventional dendritic cells (cDC1) that appear to be particularly competent in eliciting responses against dead cell antigens.18 Dendritic cells can be loaded with stressed and dying cancer cells and then be used as prophylactic or therapeutic vaccines.17 Moreover, dendritic cells can be manipulated pharmacologically to enhance their capacity to present tumor antigens to T cells.18 Such dendritic cells educate cytotoxic T lymphocytes to recognize and lyse malignant cells. Importantly, this process of T-cell-mediated cytotoxicity can elicit immunogenic cell death, hence amplifying the phenomenon and protracting the anticancer immune response.19 However, cell death affecting immune cells may play down such a desirable immunosurveillance. Specifically, it appears that dying neutrophil granulocytes produce so-called neutrophil extracellular traps (NETs) that shield cancer cells from cytotoxic immunity, hence impairing their clearance.20 Moreover, dying neutrophils can stimulate unwarranted inflammatory and autoimmune responses.21

Altogether, this volume of Immunological Reviews illustrates to which extent different cell stress and death modalities affecting malignant cells, pathogen-infected cells, or immune cells can elicit innate and cognate immune responses with vast consequences for whole-body physiology. It appears that processes that for long have been studied exclusively by cell biologists have acquired a major immunological dimension that already yields tangible impact with respect to the clinical management of malignant diseases. Future will tell whether the knowledge generated in this field will also contribute to the prevention and treatment of infectious and autoimmune diseases.

The authors declare no relevant conflicts of interest.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Immunological Reviews
Immunological Reviews 医学-免疫学
CiteScore
16.20
自引率
1.10%
发文量
118
审稿时长
4-8 weeks
期刊介绍: Immunological Reviews is a specialized journal that focuses on various aspects of immunological research. It encompasses a wide range of topics, such as clinical immunology, experimental immunology, and investigations related to allergy and the immune system. The journal follows a unique approach where each volume is dedicated solely to a specific area of immunological research. However, collectively, these volumes aim to offer an extensive and up-to-date overview of the latest advancements in basic immunology and their practical implications in clinical settings.
期刊最新文献
Immune checkpoint inhibitors in infectious disease. Pathogenic role of anti-nuclear autoantibodies in systemic sclerosis: Insights from other rheumatic diseases. The Janus (dual) model of immunoglobulin isotype evolution: Conservation and plasticity are the defining paradigms. Nanobody-based heavy chain antibodies and chimeric antibodies. Natural killer cells and engagers: Powerful weapons against cancer.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1