Innate immunity—With an adaptive twist

IF 7.5 2区 医学 Q1 IMMUNOLOGY Immunological Reviews Pub Date : 2024-04-17 DOI:10.1111/imr.13334
Steven Z. Josefowicz, Joseph C. Sun
{"title":"Innate immunity—With an adaptive twist","authors":"Steven Z. Josefowicz,&nbsp;Joseph C. Sun","doi":"10.1111/imr.13334","DOIUrl":null,"url":null,"abstract":"<p>Decades of discovery have led immunologists to compartmentalize the mammalian immune system into two components: innate and adaptive immunity. The textbooks and traditional viewpoint describe the innate immune system as rapid and non-specific, whereas the adaptive immune system consisting of T and B cells is delayed but specific and possessing memory. Every immune cell type that is not a T or B cell is broadly lumped under the umbrella of innate immunity. However, recent research has shown us that certain innate immune cells can possess features of adaptive immunity, including immunological memory.</p><p>Anecdotal evidence of memory in the innate immune system—a memory independent of T and B cell-mediated antigen-specific memory—has existed for a century or more and included observations in plants and animals, including humans. Only recently, however, have the specific cellular and molecular mechanisms started to emerge, highlighting fundamentals of immunity and previously unknown functional ‘levers’ that tune immune tone. The key cellular players, natural killer (NK) cells and myeloid cells, are found at the forefront of this paradigm-shifting revolution and central to this volume of Immunological Reviews. Two decades ago, NK cells were first shown to possess adaptive immune features from antigen specificity to clonal expansion to long-lived memory to recall responses. Next, myeloid cells were proposed to possess anamnestic responses after initial stimulation in a process termed “trained immunity.” Although our understanding of the mechanisms driving such adaptive characteristics in innate immune cells has expanded in recent years, there is still much to be learned about the important features of innate immune memory. Future studies will illuminate additional external signals inducing durable memory, cellular and metabolic processes required, underlying transcription factor and epigenetic programs and their durability, and finally the impact on health and disease.</p><p>The first group of reviews in this volume address how mouse and human NK cells respond to various environmental stimuli that program their clonality, gene expression, metabolism, effector function, survival, trafficking, tissue residency, and memory. Reviews from Delconte and Sun<span><sup>1</sup></span> and Ashkar and colleagues,<span><sup>2</sup></span> focus on underlying organ-specific metabolic mechanisms in mouse and human NK cells, respectively, in the contexts of nutrition and health versus host perturbations including fasting, infection, and cancer. Aguilar and Lanier<span><sup>3</sup></span> highlight how adaptive features of NK cells including clonal expansion depend upon specific signaling via ITAM-containing receptors. Reviews from Degli-Esposti and colleagues,<span><sup>4</sup></span> Hermans and O'Sullivan,<span><sup>5</sup></span> and Ruckert and Romagnani<span><sup>6</sup></span> focus on the selection of mouse and human NK cell clones to be epigenetically primed for effector function and survival during their response against cytomegalovirus infection, where the subsequent memory NK cells can become resident in certain tissues to protect against autoimmunity and secondary infection.</p><p>A second group of reviews centers around how innate lymphoid cells (ILC) can possess heterogeneity, plasticity, and memory following exposure to specific inflammatory signals. Colonna and colleagues<span><sup>7</sup></span> review how group 1 ILCs (ILC1) are distinct from NK cells and driven by shared and unique transcription factors to reside as sentinels embedded and resident in tissues. Martinez-Gonzalez and Takei<span><sup>8</sup></span> summarize how group 2 ILCs (ILC2) can mediate allergic recall responses following type 2 cytokine exposure and how ILC2 memory can result in both beneficial and deleterious effects on health. Serafini and Di Santo<span><sup>9</sup></span> discuss how group 3 ILCs (ILC3) can be primed in intestinal tissue to possess memory during bacteria-driven inflammation and the consequences of ILC3 priming. All reviews in this group point out that ILC1, ILC2, and ILC3 heterogeneity has been recently revealed using various single-cell sequencing approaches.</p><p>In addition to NK cells and ILCs, which lack germline-rearranged antigen receptors, innate T cells are also included in a third group of reviews that focus on mucosal-associated invariant T cells (MAIT) and tumor-associated innate T lymphocytes. Prlic and colleagues<span><sup>10</sup></span> focus on the functions of MAIT cells in healthy versus inflamed tissues and discuss the TCR and cytokine signals that this subset of innate T cells integrates when activated. Li and colleagues<span><sup>11</sup></span> describe a subset of innate T cells with cytotoxic potential that are distinct from conventional T cells but are similarly recruited to tumors and provide an important cancer immunosurveillance function.</p><p>One common feature of these lymphoid lineage innate (and innate-like) immune cells described above is their shared potential for clonal expansion, a potent and inherently memory-forming characteristic if these cells persist, especially with acquired type-specific inflammatory programs. In contrast, cells of the myeloid lineage generally lack the potential for extensive expansion. Despite this, myeloid cells have a commanding ability to initiate and control immune responses as a result of their potential to robustly initiate inflammatory cascades, recruit diverse other immune cells, and present antigen to T cells and the innate lymphoid cells. In recent years, it has become clear that these powerful immune kickstarting activities in myeloid cells are subject to durable epigenetic and metabolic tuning following inflammatory challenges. These altered myeloid phenotypes can substantively change immune function across a spectrum from bolstering defense to driving or exacerbating inflammatory pathology.</p><p>One ostensible paradox in the field has been that many of these myeloid cells that mediate durable innate immune memory are themselves short-lived. Recent studies reviewed and elaborated in this issue highlight the critical role of myeloid progenitor cells and self-renewing hematopoietic stem cells (HSC) as a major cellular reservoir of inflammatory memory capable of passing epigenetic programs to mature progeny myeloid cells. Netea and colleagues<span><sup>12</sup></span> comprehensively review these themes and also highlight important emerging areas of research in this field warranting further investigation, including further illuminating molecular mechanisms of encoding memory, potential and function of transgenerational transmission of innate memory, and the effects of existing therapies on these pathways and the potential of a new class of therapeutics that could coopt them. Sadeghi and Divangahi<span><sup>13</sup></span> describe the evolutionary and phylogenetic origins of trained immunity and the growing catalogue of adaptive features in innate immune cells and their progenitors. Josefowicz and colleagues<span><sup>14</sup></span> discuss the potency of HSC reprogramming for changing immune function, including the epigenetic priming of antigen presentation pathways in HSC and new methods that enable analysis of epigenetic reprogramming of immune progenitors in human disease using blood rather than invasive bone marrow biopsies. Neher and colleagues<span><sup>15</sup></span> review heterogeneity and plasticity of microglia and provide new analysis of single microglia combined transcriptomic and epigenomic programs, highlighting how these approaches can enhance understanding of pathophysiology involving these important central nervous system myeloid sentinels. Next, Barreiro and colleagues<span><sup>16</sup></span> synthesize the fields of population immunogenomics and epigenomics and discuss how a combination of genetic ancestry (and allelic variants) and environment tune diverse immune pathway activities with relevance to our coevolution with global pathogens and natural selection through human history.</p><p>Some cytokines play an outsized role in initiating epigenetic inflammatory memory, and a key example of this are interferon (IFN) family cytokines, both type I IFN (IFN-a/b/e/o) and type II IFN (IFN-g). Barrat and colleagues<span><sup>17</sup></span> discuss plasmacytoid dendritic cells—potent producers of type I IFN—and present new data on how these cells are regulated and their effects on inflammation and wound healing. Mishra and Ivashkiv<span><sup>18</sup></span> highlight the epigenetic regulation of inflammatory genes by IFN signaling, the central activity of STAT1 and IRF1 transcription factors, and present a helpful spectrum of training-priming effects mediated by IFNs. O'Neill and colleagues<span><sup>19</sup></span> focus on immunometabolic programs that respond to inflammation and regulate type I IFN production.</p><p>Innate immune memory can be encoded beyond immune cells, including in epithelial stem cells,<span><sup>20</sup></span> and at the tissue level.<span><sup>21, 22</sup></span> How these tissue-level inflammatory memories and adaptations develop and accumulate in early life and what environmental factors influence them is an area of recent and intensive research that may address mechanisms underlying surges in industrial-era diseases. Two reviews cover these concepts with Fernandes and Lim<span><sup>23</sup></span> focusing on maternal-immune education in offspring, and Iza and Brown<span><sup>24</sup></span> discussing early life imprinting of intestinal immune tolerance and the key role of unconventional antigen-presenting cells.</p><p>In conclusion, our new and expanding understanding that cellular components of the innate immune system can possess memory will cause us to re-evaluate vaccine strategies that have traditionally targeted T or B cells. Furthermore, because NK cells, ILCs, and myeloid cells can possess anamnestic responses against pathogens, allergens, tumors, and other inflammatory stimuli, new considerations must be taken in how we assess long-lived immune responses following initial exposure to various insults. This is an exciting time in immunology as we seek to more deeply understanding the cellular and molecular mechanisms that underlie generation and maintenance of immunological memory in the context of innate immune system.</p><p>SZJ is co-founder of Epistemyx Inc.</p>","PeriodicalId":178,"journal":{"name":"Immunological Reviews","volume":null,"pages":null},"PeriodicalIF":7.5000,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imr.13334","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Immunological Reviews","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1111/imr.13334","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

Abstract

Decades of discovery have led immunologists to compartmentalize the mammalian immune system into two components: innate and adaptive immunity. The textbooks and traditional viewpoint describe the innate immune system as rapid and non-specific, whereas the adaptive immune system consisting of T and B cells is delayed but specific and possessing memory. Every immune cell type that is not a T or B cell is broadly lumped under the umbrella of innate immunity. However, recent research has shown us that certain innate immune cells can possess features of adaptive immunity, including immunological memory.

Anecdotal evidence of memory in the innate immune system—a memory independent of T and B cell-mediated antigen-specific memory—has existed for a century or more and included observations in plants and animals, including humans. Only recently, however, have the specific cellular and molecular mechanisms started to emerge, highlighting fundamentals of immunity and previously unknown functional ‘levers’ that tune immune tone. The key cellular players, natural killer (NK) cells and myeloid cells, are found at the forefront of this paradigm-shifting revolution and central to this volume of Immunological Reviews. Two decades ago, NK cells were first shown to possess adaptive immune features from antigen specificity to clonal expansion to long-lived memory to recall responses. Next, myeloid cells were proposed to possess anamnestic responses after initial stimulation in a process termed “trained immunity.” Although our understanding of the mechanisms driving such adaptive characteristics in innate immune cells has expanded in recent years, there is still much to be learned about the important features of innate immune memory. Future studies will illuminate additional external signals inducing durable memory, cellular and metabolic processes required, underlying transcription factor and epigenetic programs and their durability, and finally the impact on health and disease.

The first group of reviews in this volume address how mouse and human NK cells respond to various environmental stimuli that program their clonality, gene expression, metabolism, effector function, survival, trafficking, tissue residency, and memory. Reviews from Delconte and Sun1 and Ashkar and colleagues,2 focus on underlying organ-specific metabolic mechanisms in mouse and human NK cells, respectively, in the contexts of nutrition and health versus host perturbations including fasting, infection, and cancer. Aguilar and Lanier3 highlight how adaptive features of NK cells including clonal expansion depend upon specific signaling via ITAM-containing receptors. Reviews from Degli-Esposti and colleagues,4 Hermans and O'Sullivan,5 and Ruckert and Romagnani6 focus on the selection of mouse and human NK cell clones to be epigenetically primed for effector function and survival during their response against cytomegalovirus infection, where the subsequent memory NK cells can become resident in certain tissues to protect against autoimmunity and secondary infection.

A second group of reviews centers around how innate lymphoid cells (ILC) can possess heterogeneity, plasticity, and memory following exposure to specific inflammatory signals. Colonna and colleagues7 review how group 1 ILCs (ILC1) are distinct from NK cells and driven by shared and unique transcription factors to reside as sentinels embedded and resident in tissues. Martinez-Gonzalez and Takei8 summarize how group 2 ILCs (ILC2) can mediate allergic recall responses following type 2 cytokine exposure and how ILC2 memory can result in both beneficial and deleterious effects on health. Serafini and Di Santo9 discuss how group 3 ILCs (ILC3) can be primed in intestinal tissue to possess memory during bacteria-driven inflammation and the consequences of ILC3 priming. All reviews in this group point out that ILC1, ILC2, and ILC3 heterogeneity has been recently revealed using various single-cell sequencing approaches.

In addition to NK cells and ILCs, which lack germline-rearranged antigen receptors, innate T cells are also included in a third group of reviews that focus on mucosal-associated invariant T cells (MAIT) and tumor-associated innate T lymphocytes. Prlic and colleagues10 focus on the functions of MAIT cells in healthy versus inflamed tissues and discuss the TCR and cytokine signals that this subset of innate T cells integrates when activated. Li and colleagues11 describe a subset of innate T cells with cytotoxic potential that are distinct from conventional T cells but are similarly recruited to tumors and provide an important cancer immunosurveillance function.

One common feature of these lymphoid lineage innate (and innate-like) immune cells described above is their shared potential for clonal expansion, a potent and inherently memory-forming characteristic if these cells persist, especially with acquired type-specific inflammatory programs. In contrast, cells of the myeloid lineage generally lack the potential for extensive expansion. Despite this, myeloid cells have a commanding ability to initiate and control immune responses as a result of their potential to robustly initiate inflammatory cascades, recruit diverse other immune cells, and present antigen to T cells and the innate lymphoid cells. In recent years, it has become clear that these powerful immune kickstarting activities in myeloid cells are subject to durable epigenetic and metabolic tuning following inflammatory challenges. These altered myeloid phenotypes can substantively change immune function across a spectrum from bolstering defense to driving or exacerbating inflammatory pathology.

One ostensible paradox in the field has been that many of these myeloid cells that mediate durable innate immune memory are themselves short-lived. Recent studies reviewed and elaborated in this issue highlight the critical role of myeloid progenitor cells and self-renewing hematopoietic stem cells (HSC) as a major cellular reservoir of inflammatory memory capable of passing epigenetic programs to mature progeny myeloid cells. Netea and colleagues12 comprehensively review these themes and also highlight important emerging areas of research in this field warranting further investigation, including further illuminating molecular mechanisms of encoding memory, potential and function of transgenerational transmission of innate memory, and the effects of existing therapies on these pathways and the potential of a new class of therapeutics that could coopt them. Sadeghi and Divangahi13 describe the evolutionary and phylogenetic origins of trained immunity and the growing catalogue of adaptive features in innate immune cells and their progenitors. Josefowicz and colleagues14 discuss the potency of HSC reprogramming for changing immune function, including the epigenetic priming of antigen presentation pathways in HSC and new methods that enable analysis of epigenetic reprogramming of immune progenitors in human disease using blood rather than invasive bone marrow biopsies. Neher and colleagues15 review heterogeneity and plasticity of microglia and provide new analysis of single microglia combined transcriptomic and epigenomic programs, highlighting how these approaches can enhance understanding of pathophysiology involving these important central nervous system myeloid sentinels. Next, Barreiro and colleagues16 synthesize the fields of population immunogenomics and epigenomics and discuss how a combination of genetic ancestry (and allelic variants) and environment tune diverse immune pathway activities with relevance to our coevolution with global pathogens and natural selection through human history.

Some cytokines play an outsized role in initiating epigenetic inflammatory memory, and a key example of this are interferon (IFN) family cytokines, both type I IFN (IFN-a/b/e/o) and type II IFN (IFN-g). Barrat and colleagues17 discuss plasmacytoid dendritic cells—potent producers of type I IFN—and present new data on how these cells are regulated and their effects on inflammation and wound healing. Mishra and Ivashkiv18 highlight the epigenetic regulation of inflammatory genes by IFN signaling, the central activity of STAT1 and IRF1 transcription factors, and present a helpful spectrum of training-priming effects mediated by IFNs. O'Neill and colleagues19 focus on immunometabolic programs that respond to inflammation and regulate type I IFN production.

Innate immune memory can be encoded beyond immune cells, including in epithelial stem cells,20 and at the tissue level.21, 22 How these tissue-level inflammatory memories and adaptations develop and accumulate in early life and what environmental factors influence them is an area of recent and intensive research that may address mechanisms underlying surges in industrial-era diseases. Two reviews cover these concepts with Fernandes and Lim23 focusing on maternal-immune education in offspring, and Iza and Brown24 discussing early life imprinting of intestinal immune tolerance and the key role of unconventional antigen-presenting cells.

In conclusion, our new and expanding understanding that cellular components of the innate immune system can possess memory will cause us to re-evaluate vaccine strategies that have traditionally targeted T or B cells. Furthermore, because NK cells, ILCs, and myeloid cells can possess anamnestic responses against pathogens, allergens, tumors, and other inflammatory stimuli, new considerations must be taken in how we assess long-lived immune responses following initial exposure to various insults. This is an exciting time in immunology as we seek to more deeply understanding the cellular and molecular mechanisms that underlie generation and maintenance of immunological memory in the context of innate immune system.

SZJ is co-founder of Epistemyx Inc.

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
先天性免疫--适应性变化
数十年的研究发现,免疫学家将哺乳动物的免疫系统分为两个部分:先天性免疫和适应性免疫。教科书和传统观点将先天性免疫系统描述为快速和非特异性的,而由 T 细胞和 B 细胞组成的适应性免疫系统则是延迟的,但具有特异性和记忆性。所有非 T 细胞或 B 细胞的免疫细胞类型都被笼统地归入先天性免疫的范畴。先天性免疫系统记忆的轶事证据--一种独立于 T 细胞和 B 细胞介导的抗原特异性记忆--已经存在了一个多世纪,包括对植物和动物(包括人类)的观察。然而,直到最近,具体的细胞和分子机制才开始浮出水面,凸显了免疫的基本原理和以前未知的调节免疫调节的功能 "杠杆"。自然杀伤(NK)细胞和类髓鞘细胞是这一范式转变革命的关键细胞角色,也是本卷《免疫学评论》的核心内容。二十年前,NK 细胞首次被证明具有适应性免疫特征,包括抗原特异性、克隆扩增、长效记忆和回忆反应。随后,髓系细胞被认为在最初的刺激后具有拟态反应,这一过程被称为 "训练有素的免疫"。尽管近年来我们对先天性免疫细胞这种适应性特征的驱动机制有了进一步的了解,但对于先天性免疫记忆的重要特征,我们仍有许多东西需要学习。未来的研究将揭示诱导持久记忆的更多外部信号、所需的细胞和代谢过程、潜在的转录因子和表观遗传程序及其持久性,以及最终对健康和疾病的影响。本卷的第一组综述探讨了小鼠和人类 NK 细胞如何对各种环境刺激做出反应,从而对其克隆性、基因表达、新陈代谢、效应功能、存活、贩运、组织驻留和记忆进行编程。Delconte 和 Sun1 以及 Ashkar 及其同事2 的综述分别侧重于小鼠和人类 NK 细胞在营养和健康与宿主干扰(包括禁食、感染和癌症)背景下的潜在器官特异性代谢机制。Aguilar 和 Lanier3 强调了 NK 细胞的适应性特征(包括克隆扩增)如何依赖于通过含 ITAM 受体发出的特定信号。Degli-Esposti及其同事4、Hermans和O'Sullivan5以及Ruckert和Romagnani6的综述重点介绍了小鼠和人类NK细胞克隆的选择,这些克隆在对抗巨细胞病毒感染的过程中被表观遗传学地激活了效应功能和存活能力,随后的记忆性NK细胞可以驻留在某些组织中,防止自身免疫和二次感染。第二组综述围绕先天性淋巴细胞(ILC)如何在暴露于特定炎症信号后具有异质性、可塑性和记忆性。Colonna 及其同事7 综述了第 1 组 ILC(ILC1)如何区别于 NK 细胞,并在共享和独特转录因子的驱动下作为哨兵嵌入和驻留在组织中。Martinez-Gonzalez 和 Takei8 总结了第 2 组 ILC(ILC2)如何在暴露于 2 型细胞因子后介导过敏性回忆反应,以及 ILC2 记忆如何对健康产生有益和有害的影响。Serafini 和 Di Santo9 讨论了第 3 组 ILCs(ILC3)如何在细菌驱动的炎症过程中在肠道组织中形成记忆,以及 ILC3 形成记忆的后果。除了缺乏种系重组抗原受体的 NK 细胞和 ILCs 外,先天性 T 细胞也包括在第三组综述中,这组综述的重点是粘膜相关不变 T 细胞(MAIT)和肿瘤相关先天性 T 淋巴细胞。Prlic 及其同事10 重点研究了 MAIT 细胞在健康组织和炎症组织中的功能,并讨论了这一先天性 T 细胞亚群在激活时整合的 TCR 和细胞因子信号。Li及其同事11描述了具有细胞毒性潜能的先天性T细胞亚群,它们与传统的T细胞不同,但同样会被招募到肿瘤中,并提供重要的癌症免疫监视功能。上述这些淋巴系先天性(和类先天性)免疫细胞的一个共同特征是它们都具有克隆扩增的潜能,如果这些细胞持续存在,尤其是在获得性特异性炎症程序的情况下,这种潜能就会形成记忆。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 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