首页 > 最新文献

Trends in Immunology最新文献

英文 中文
Decoding brain border immunity to enable future therapeutic avenues. 破解脑边界免疫,为未来的治疗提供途径。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-08 DOI: 10.1016/j.it.2025.12.007
Lien Van Hoecke, Lore Van Acker, Roosmarijn E Vandenbroucke

The brain is no longer viewed as immunologically isolated but as an organ surrounded by dynamic border compartments that coordinate surveillance, drainage, and communication with the periphery. Key interfaces - including the meninges, blood-brain barrier, choroid plexus (ChP), and skull bone marrow - host specialized immune niches that regulate antigen sampling, leukocyte trafficking, and neuroimmune signaling. Recent advances in imaging and in single-cell and spatial profiling have revealed previously unrecognized cell types, migration routes, and barrier specializations that shape central nervous system (CNS) immunity in health and disease. Understanding how these border tissues sense, integrate, and modulate immune activity opens opportunities for therapeutically tuning neuroimmune responses at the brain's periphery while preserving parenchymal integrity.

大脑不再被视为免疫隔离的器官,而是一个被动态边界隔间包围的器官,这些隔间协调监视、排水和与周围的交流。关键的界面-包括脑膜、血脑屏障、脉络膜丛(ChP)和颅骨骨髓-宿主专门的免疫龛,调节抗原取样、白细胞运输和神经免疫信号。成像、单细胞和空间分析的最新进展揭示了以前未被识别的细胞类型、迁移路线和屏障特化,这些细胞类型、迁移路线和屏障特化在健康和疾病中塑造了中枢神经系统(CNS)免疫。了解这些边界组织如何感知、整合和调节免疫活动,为在保持脑实质完整性的同时治疗性地调整脑周围的神经免疫反应提供了机会。
{"title":"Decoding brain border immunity to enable future therapeutic avenues.","authors":"Lien Van Hoecke, Lore Van Acker, Roosmarijn E Vandenbroucke","doi":"10.1016/j.it.2025.12.007","DOIUrl":"https://doi.org/10.1016/j.it.2025.12.007","url":null,"abstract":"<p><p>The brain is no longer viewed as immunologically isolated but as an organ surrounded by dynamic border compartments that coordinate surveillance, drainage, and communication with the periphery. Key interfaces - including the meninges, blood-brain barrier, choroid plexus (ChP), and skull bone marrow - host specialized immune niches that regulate antigen sampling, leukocyte trafficking, and neuroimmune signaling. Recent advances in imaging and in single-cell and spatial profiling have revealed previously unrecognized cell types, migration routes, and barrier specializations that shape central nervous system (CNS) immunity in health and disease. Understanding how these border tissues sense, integrate, and modulate immune activity opens opportunities for therapeutically tuning neuroimmune responses at the brain's periphery while preserving parenchymal integrity.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":""},"PeriodicalIF":13.9,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145946550","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Microglia in systemic neuroimmune communication: functions beyond phagocytosis. 全身神经免疫通讯中的小胶质细胞:吞噬作用以外的功能。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-07 DOI: 10.1016/j.it.2025.12.002
Anabella Ayelen Di Pietro, Sharon Powley, Elisa Perciballi, Meredith Stewart, Martine Therrien, Nicole Scott-Hewitt

Bidirectional crosstalk between the immune and nervous systems, via 'neuroimmune circuits', regulates homeostatic and inflammatory responses essential for health. Microglia, long-lived brain macrophages, act as key hubs integrating immune signals into coordinated brain responses by shifting into distinct functional states in response to local and systemic cues. In this review, we focus on how environmental signals shape these microglial states, how microglia influence other brain cells through both direct and indirect mechanisms, and emerging evidence of how microglia are impacted by, and respond to, peripheral changes. We highlight microglia as central players in systemic neuroimmune communication, influencing both brain and peripheral health, while outlining recent tools and key knowledge gaps to guide future research into mechanisms of neuroimmune circuit communication.

通过“神经免疫回路”,免疫系统和神经系统之间的双向串扰调节对健康至关重要的稳态和炎症反应。小胶质细胞是一种长寿的脑巨噬细胞,它是将免疫信号整合到协调的大脑反应中的关键枢纽,通过对局部和系统信号的反应转变为不同的功能状态。在这篇综述中,我们关注环境信号如何塑造这些小胶质细胞状态,小胶质细胞如何通过直接和间接机制影响其他脑细胞,以及小胶质细胞如何受到外周变化的影响并对其做出反应的新证据。我们强调小胶质细胞是系统性神经免疫通讯的核心参与者,影响大脑和外周健康,同时概述了最近的工具和关键知识空白,以指导未来神经免疫回路通讯机制的研究。
{"title":"Microglia in systemic neuroimmune communication: functions beyond phagocytosis.","authors":"Anabella Ayelen Di Pietro, Sharon Powley, Elisa Perciballi, Meredith Stewart, Martine Therrien, Nicole Scott-Hewitt","doi":"10.1016/j.it.2025.12.002","DOIUrl":"https://doi.org/10.1016/j.it.2025.12.002","url":null,"abstract":"<p><p>Bidirectional crosstalk between the immune and nervous systems, via 'neuroimmune circuits', regulates homeostatic and inflammatory responses essential for health. Microglia, long-lived brain macrophages, act as key hubs integrating immune signals into coordinated brain responses by shifting into distinct functional states in response to local and systemic cues. In this review, we focus on how environmental signals shape these microglial states, how microglia influence other brain cells through both direct and indirect mechanisms, and emerging evidence of how microglia are impacted by, and respond to, peripheral changes. We highlight microglia as central players in systemic neuroimmune communication, influencing both brain and peripheral health, while outlining recent tools and key knowledge gaps to guide future research into mechanisms of neuroimmune circuit communication.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":""},"PeriodicalIF":13.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145936426","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Resident T cells in the healthy and multiple sclerosis brain. 健康和多发性硬化症大脑中的常驻T细胞。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-07 DOI: 10.1016/j.it.2025.12.006
Joost Smolders, Cheng-Chih Hsiao, Inge Huitinga, Jörg Hamann

Insights into T-cell biology in the central nervous system (CNS) have evolved from early neuroinflammatory models demonstrating the pathogenic potential of autoreactive T cells to recent human studies defining resident T-cell populations in the healthy and diseased brain. We here discuss advances in postmortem brain tissue processing, flow cytometry, and transcriptional profiling revealing that human brain CD8+ and CD4+ T cells are tissue-resident memory T cells with distinct phenotypes shaped by CNS borders and parenchymal niches. These findings refine our understanding of CNS immune surveillance and provide a framework for dissecting T-cell contributions to multiple sclerosis.

对中枢神经系统(CNS)中T细胞生物学的认识已经从早期的神经炎症模型发展到最近的人类研究,证明了自身反应性T细胞的致病潜力,定义了健康和患病大脑中的常驻T细胞群。我们在这里讨论了死后脑组织处理、流式细胞术和转录谱的进展,揭示了人脑CD8+和CD4+ T细胞是组织驻留记忆T细胞,具有不同的表型,由中枢神经系统边界和实质壁龛塑造。这些发现完善了我们对中枢神经系统免疫监测的理解,并为剖析t细胞对多发性硬化症的贡献提供了一个框架。
{"title":"Resident T cells in the healthy and multiple sclerosis brain.","authors":"Joost Smolders, Cheng-Chih Hsiao, Inge Huitinga, Jörg Hamann","doi":"10.1016/j.it.2025.12.006","DOIUrl":"https://doi.org/10.1016/j.it.2025.12.006","url":null,"abstract":"<p><p>Insights into T-cell biology in the central nervous system (CNS) have evolved from early neuroinflammatory models demonstrating the pathogenic potential of autoreactive T cells to recent human studies defining resident T-cell populations in the healthy and diseased brain. We here discuss advances in postmortem brain tissue processing, flow cytometry, and transcriptional profiling revealing that human brain CD8<sup>+</sup> and CD4<sup>+</sup> T cells are tissue-resident memory T cells with distinct phenotypes shaped by CNS borders and parenchymal niches. These findings refine our understanding of CNS immune surveillance and provide a framework for dissecting T-cell contributions to multiple sclerosis.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":""},"PeriodicalIF":13.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145936544","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tissue-resident regulatory T cells: modulators of local immunity. 组织常驻调节性T细胞:局部免疫的调节剂。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-07 DOI: 10.1016/j.it.2025.12.001
Yun Shi, Lingling Wang, Chuntong Bao, Guanpeng Wang, Zuoming Sun

Regulatory T cells (Tregs) have gained renewed attention for their diverse roles beyond immune suppression. This review integrates recent discoveries on how tissue-resident Tregs integrate immune, metabolic, and neural cues to maintain organ homeostasis and regeneration. Across adipose tissue, intestine, brain, and skin, Tregs coordinate local networks that couple immune tolerance with metabolic balance and tissue repair. We further discuss therapeutic advances - including antigen-specific chimeric antigen receptor (CAR)/T cell receptor (TCR) Tregs, interleukin 2 (IL-2) muteins, and metabolic modulation - that aim to harness Tregs for treating autoimmunity and chronic inflammation. Together, these insights highlight Tregs as central interpreters of tissue context and as promising targets for next-generation precision immunotherapy.

调节性T细胞(Tregs)因其在免疫抑制之外的多种作用而重新受到关注。这篇综述整合了最近关于组织内treg如何整合免疫、代谢和神经信号来维持器官稳态和再生的发现。在脂肪组织、肠道、大脑和皮肤中,Tregs协调局部网络,将免疫耐受与代谢平衡和组织修复结合起来。我们进一步讨论了治疗进展-包括抗原特异性嵌合抗原受体(CAR)/T细胞受体(TCR) Tregs,白细胞介素2 (IL-2)突变蛋白和代谢调节-旨在利用Tregs治疗自身免疫和慢性炎症。总之,这些见解突出了Tregs作为组织背景的核心解释者和下一代精确免疫治疗的有希望的靶点。
{"title":"Tissue-resident regulatory T cells: modulators of local immunity.","authors":"Yun Shi, Lingling Wang, Chuntong Bao, Guanpeng Wang, Zuoming Sun","doi":"10.1016/j.it.2025.12.001","DOIUrl":"https://doi.org/10.1016/j.it.2025.12.001","url":null,"abstract":"<p><p>Regulatory T cells (T<sub>reg</sub><sub>s</sub>) have gained renewed attention for their diverse roles beyond immune suppression. This review integrates recent discoveries on how tissue-resident T<sub>regs</sub> integrate immune, metabolic, and neural cues to maintain organ homeostasis and regeneration. Across adipose tissue, intestine, brain, and skin, T<sub>regs</sub> coordinate local networks that couple immune tolerance with metabolic balance and tissue repair. We further discuss therapeutic advances - including antigen-specific chimeric antigen receptor (CAR)/T cell receptor (TCR) T<sub>regs</sub>, interleukin 2 (IL-2) muteins, and metabolic modulation - that aim to harness T<sub>regs</sub> for treating autoimmunity and chronic inflammation. Together, these insights highlight T<sub>regs</sub> as central interpreters of tissue context and as promising targets for next-generation precision immunotherapy.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":""},"PeriodicalIF":13.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Interleukin-23 biology linking mucosal immunity to autoimmune diseases and cancer. 白细胞介素-23生物学将粘膜免疫与自身免疫性疾病和癌症联系起来。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-03 DOI: 10.1016/j.it.2025.12.005
Anees Ahmed, Gregory F Sonnenberg

Interleukin-23 (IL-23) is a pleiotropic cytokine that maintains the delicate balance between tolerance to commensal microbiota and defense against pathogens at mucosal barriers. When dysregulated, IL-23 becomes a key driver of chronic inflammation, with therapeutics blocking this pathway being successfully harnessed in the clinic. In this review, we discuss recently uncovered biology of IL-23 in the context of mucosal immunity, which intimately links the role of this pathway to the pathophysiology of autoimmunity, chronic inflammation, and emerging functions in cancer. Through the lens of the cell types that respond to IL-23, the engaged effector programs, and the key functions in health and disease, we highlight recent advances and opportunities to better understand the dichotomous outcomes mediated by this cytokine.

白细胞介素-23 (IL-23)是一种多效性细胞因子,维持对共生微生物群的耐受性和粘膜屏障对病原体的防御之间的微妙平衡。当失调时,IL-23成为慢性炎症的关键驱动因素,治疗方法阻断这一途径已在临床中成功利用。在这篇综述中,我们讨论了最近发现的IL-23在粘膜免疫中的生物学作用,并将其与自身免疫、慢性炎症和癌症中新出现的功能的病理生理作用密切联系起来。通过对IL-23作出反应的细胞类型、参与的效应程序以及健康和疾病中的关键功能,我们强调了最近的进展和机会,以更好地理解由该细胞因子介导的二分结果。
{"title":"Interleukin-23 biology linking mucosal immunity to autoimmune diseases and cancer.","authors":"Anees Ahmed, Gregory F Sonnenberg","doi":"10.1016/j.it.2025.12.005","DOIUrl":"https://doi.org/10.1016/j.it.2025.12.005","url":null,"abstract":"<p><p>Interleukin-23 (IL-23) is a pleiotropic cytokine that maintains the delicate balance between tolerance to commensal microbiota and defense against pathogens at mucosal barriers. When dysregulated, IL-23 becomes a key driver of chronic inflammation, with therapeutics blocking this pathway being successfully harnessed in the clinic. In this review, we discuss recently uncovered biology of IL-23 in the context of mucosal immunity, which intimately links the role of this pathway to the pathophysiology of autoimmunity, chronic inflammation, and emerging functions in cancer. Through the lens of the cell types that respond to IL-23, the engaged effector programs, and the key functions in health and disease, we highlight recent advances and opportunities to better understand the dichotomous outcomes mediated by this cytokine.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":""},"PeriodicalIF":13.9,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145897087","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DNA methylation and histone modifications drive the trained immunity duration. DNA甲基化和组蛋白修饰驱动训练的免疫持续时间。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-01 Epub Date: 2025-11-06 DOI: 10.1016/j.it.2025.10.004
Mohua Liu, Xihui Shen, Lei Xu

Trained immunity (TRIM) is a de facto form of innate immune memory. While histone modifications contribute to TRIM, their reversible nature and susceptibility to dilution during cell division cannot fully account for its long-term persistence. Here, we propose that DNA methylation patterns, particularly hypomethylation at proinflammatory gene loci, could serve as a key epigenetic mechanism contributing to long-term TRIM. Mechanistically, these hypomethylated states are biochemically stable and faithfully inherited through cell division, acting as a permissive scaffold that enables the rapid accumulation of activating histone marks upon restimulation. This DNA-methylation-mediated process could underpin the durability of TRIM across multiple contexts, including hematopoietic stem cell self-renewal, differentiation from central to peripheral compartments, and autonomy of tissue-resident cells.

训练免疫(TRIM)实际上是先天免疫记忆的一种形式。虽然组蛋白修饰有助于TRIM,但它们的可逆性和细胞分裂过程中稀释的易感性并不能完全解释其长期持久性。在这里,我们提出DNA甲基化模式,特别是促炎基因位点的低甲基化,可能是导致长期TRIM的关键表观遗传机制。从机制上讲,这些低甲基化状态是生物化学稳定的,并通过细胞分裂忠实地遗传,作为一个允许的支架,使激活组蛋白标记在再刺激时快速积累。这种dna甲基化介导的过程可以支持TRIM在多种情况下的持久性,包括造血干细胞自我更新,从中央到外周区室的分化以及组织驻留细胞的自主性。
{"title":"DNA methylation and histone modifications drive the trained immunity duration.","authors":"Mohua Liu, Xihui Shen, Lei Xu","doi":"10.1016/j.it.2025.10.004","DOIUrl":"10.1016/j.it.2025.10.004","url":null,"abstract":"<p><p>Trained immunity (TRIM) is a de facto form of innate immune memory. While histone modifications contribute to TRIM, their reversible nature and susceptibility to dilution during cell division cannot fully account for its long-term persistence. Here, we propose that DNA methylation patterns, particularly hypomethylation at proinflammatory gene loci, could serve as a key epigenetic mechanism contributing to long-term TRIM. Mechanistically, these hypomethylated states are biochemically stable and faithfully inherited through cell division, acting as a permissive scaffold that enables the rapid accumulation of activating histone marks upon restimulation. This DNA-methylation-mediated process could underpin the durability of TRIM across multiple contexts, including hematopoietic stem cell self-renewal, differentiation from central to peripheral compartments, and autonomy of tissue-resident cells.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":"61-76"},"PeriodicalIF":13.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145472298","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Germinal centre B cell disruption by non-typhoidal Salmonella. 生发中心B细胞被非伤寒沙门氏菌破坏。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-01 Epub Date: 2025-10-03 DOI: 10.1016/j.it.2025.09.007
Francisco Osorio-Barrios, Siegfried Hapfelmeier

Salmonella enterica serovar Typhimurium (STm) represents a major global health burden. Strains endemic in sub-Saharan Africa cause life-threatening invasive non-typhoidal salmonellosis (iNTS) in vulnerable populations. Studies in the iNTS-like mouse model show that STm induces profound germinal centre (GC) disruption, impairing high-affinity, long-lived antibody and memory B cell formation - affecting nascent and pre-existing GC reactions. Lipopolysaccharide (LPS) and specific STm type 3 secretion effectors drive GC collapse, but the determining bacteria-host interactions are still unclear. Although STm induces an extrafollicular (EF) B cell response generating protective antibodies, their longevity remains unclear. With no licensed human vaccine for iNTS, we propose that vaccine strategies should consider ways to protect GC integrity and include GC parameters as endpoints in preclinical trials.

肠炎沙门氏菌血清型鼠伤寒沙门氏菌(STm)是一种主要的全球健康负担。撒哈拉以南非洲流行的菌株在脆弱人群中引起危及生命的侵袭性非伤寒沙门氏菌病(iNTS)。在ts样小鼠模型中的研究表明,STm诱导生发中心(GC)严重破坏,损害高亲和力,长寿命抗体和记忆B细胞的形成-影响新生和预先存在的GC反应。脂多糖(LPS)和特定的STm 3型分泌效应物驱动GC崩溃,但决定细菌与宿主的相互作用尚不清楚。虽然STm诱导滤泡外(EF) B细胞反应产生保护性抗体,但其寿命尚不清楚。由于没有获得许可的iNTS人疫苗,我们建议疫苗策略应考虑保护GC完整性的方法,并将GC参数作为临床前试验的终点。
{"title":"Germinal centre B cell disruption by non-typhoidal Salmonella.","authors":"Francisco Osorio-Barrios, Siegfried Hapfelmeier","doi":"10.1016/j.it.2025.09.007","DOIUrl":"10.1016/j.it.2025.09.007","url":null,"abstract":"<p><p>Salmonella enterica serovar Typhimurium (STm) represents a major global health burden. Strains endemic in sub-Saharan Africa cause life-threatening invasive non-typhoidal salmonellosis (iNTS) in vulnerable populations. Studies in the iNTS-like mouse model show that STm induces profound germinal centre (GC) disruption, impairing high-affinity, long-lived antibody and memory B cell formation - affecting nascent and pre-existing GC reactions. Lipopolysaccharide (LPS) and specific STm type 3 secretion effectors drive GC collapse, but the determining bacteria-host interactions are still unclear. Although STm induces an extrafollicular (EF) B cell response generating protective antibodies, their longevity remains unclear. With no licensed human vaccine for iNTS, we propose that vaccine strategies should consider ways to protect GC integrity and include GC parameters as endpoints in preclinical trials.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":"19-28"},"PeriodicalIF":13.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145228666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Coming of age: mRNA vaccines for orthoflaviviruses. 成年期:正黄病毒mRNA疫苗
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-01 Epub Date: 2025-11-06 DOI: 10.1016/j.it.2025.10.005
Fahima Akther, Norbert Pardi, David R Martinez

Orthoflaviviruses - including dengue, Zika, yellow fever, Japanese encephalitis, and Powassan viruses - are mosquito- and tick-borne members of the family Flaviviridae. Orthoflaviviruses pose major public health threats, with the potential for epidemics and pandemics. Lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA vaccines offer a powerful platform by delivering in vitro-synthesized viral antigen-encoding mRNAs into the host, where they generate proteins that trigger robust immune responses. These synthetic platforms simplify the expression of complex viral glycoproteins, allow rapid and scalable manufacturing that is critical in a pandemic/epidemic scenario, and support multivalent designs to broaden protection. This review highlights recent advancements in mRNA vaccines for orthoflaviviruses and examines how innovations in antigen design and delivery platforms may offer broad, safe, and durable protection against diverse pathogenic orthoflaviviruses.

正黄病毒——包括登革热、寨卡病毒、黄热病、日本脑炎和波瓦桑病毒——是黄病毒科的蚊媒和蜱媒成员。正黄病毒对公共卫生构成重大威胁,有可能造成流行病和大流行。脂质纳米颗粒(LNP)封装的核苷修饰mRNA疫苗提供了一个强大的平台,通过将体外合成的病毒抗原编码mRNA递送到宿主体内,在宿主体内产生触发强大免疫反应的蛋白质。这些合成平台简化了复杂病毒糖蛋白的表达,允许快速和可扩展的制造,这在大流行/流行病的情况下至关重要,并支持多价设计以扩大保护。本综述重点介绍了针对正黄病毒的mRNA疫苗的最新进展,并探讨了抗原设计和递送平台的创新如何提供广泛、安全和持久的保护,以对抗多种致病性正黄病毒。
{"title":"Coming of age: mRNA vaccines for orthoflaviviruses.","authors":"Fahima Akther, Norbert Pardi, David R Martinez","doi":"10.1016/j.it.2025.10.005","DOIUrl":"10.1016/j.it.2025.10.005","url":null,"abstract":"<p><p>Orthoflaviviruses - including dengue, Zika, yellow fever, Japanese encephalitis, and Powassan viruses - are mosquito- and tick-borne members of the family Flaviviridae. Orthoflaviviruses pose major public health threats, with the potential for epidemics and pandemics. Lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA vaccines offer a powerful platform by delivering in vitro-synthesized viral antigen-encoding mRNAs into the host, where they generate proteins that trigger robust immune responses. These synthetic platforms simplify the expression of complex viral glycoproteins, allow rapid and scalable manufacturing that is critical in a pandemic/epidemic scenario, and support multivalent designs to broaden protection. This review highlights recent advancements in mRNA vaccines for orthoflaviviruses and examines how innovations in antigen design and delivery platforms may offer broad, safe, and durable protection against diverse pathogenic orthoflaviviruses.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":"43-60"},"PeriodicalIF":13.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145460517","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Beyond the innate immune system: rethinking inflammasomes in multiple sclerosis. 超越先天免疫系统:重新思考多发性硬化症中的炎性体。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-01 Epub Date: 2025-11-20 DOI: 10.1016/j.it.2025.10.014
Gayel Duran, Janne Verreycken, Yvonne Dombrowski, Mohamed Lamkanfi, Paulien Baeten, Bieke Broux

Inflammasomes have emerged as central regulators of (auto)immune pathology, including multiple sclerosis (MS). Once exclusively considered in the domain of myeloid cells, both canonical and noncanonical inflammasomes are now recognized in diverse immune and nonimmune populations relevant to MS, including T lymphocytes, blood-brain barrier (BBB) endothelial cells (EnC), and oligodendrocytes (ODCs). Elevated inflammasome activity is evident in patient-derived samples, particularly within active brain lesions. Experimental autoimmune encephalomyelitis (EAE) models confirm the pathogenic contribution of inflammasomes, as genetic deletion or pharmacological inhibition of inflammasomes mitigate disease. These advances position inflammasomes at the intersection of neuroinflammation and neurodegeneration, and highlight inflammasome inhibition as a promising therapeutic avenue currently under investigation in preclinical and early clinical studies.

炎性小体已成为(自身)免疫病理的中枢调节因子,包括多发性硬化症(MS)。曾经被认为只存在于髓系细胞领域的典型和非典型炎性小体,现在在与MS相关的多种免疫和非免疫群体中都被发现,包括T淋巴细胞、血脑屏障(BBB)内皮细胞(EnC)和少突胶质细胞(ODCs)。在患者来源的样本中,炎症小体活性明显升高,特别是在活动性脑病变中。实验性自身免疫性脑脊髓炎(EAE)模型证实了炎症小体的致病作用,因为炎症小体的基因缺失或药理抑制可减轻疾病。这些进展将炎症小体定位于神经炎症和神经退行性变的交叉点,并强调炎症小体抑制是目前正在临床前和早期临床研究中研究的有前途的治疗途径。
{"title":"Beyond the innate immune system: rethinking inflammasomes in multiple sclerosis.","authors":"Gayel Duran, Janne Verreycken, Yvonne Dombrowski, Mohamed Lamkanfi, Paulien Baeten, Bieke Broux","doi":"10.1016/j.it.2025.10.014","DOIUrl":"10.1016/j.it.2025.10.014","url":null,"abstract":"<p><p>Inflammasomes have emerged as central regulators of (auto)immune pathology, including multiple sclerosis (MS). Once exclusively considered in the domain of myeloid cells, both canonical and noncanonical inflammasomes are now recognized in diverse immune and nonimmune populations relevant to MS, including T lymphocytes, blood-brain barrier (BBB) endothelial cells (EnC), and oligodendrocytes (ODCs). Elevated inflammasome activity is evident in patient-derived samples, particularly within active brain lesions. Experimental autoimmune encephalomyelitis (EAE) models confirm the pathogenic contribution of inflammasomes, as genetic deletion or pharmacological inhibition of inflammasomes mitigate disease. These advances position inflammasomes at the intersection of neuroinflammation and neurodegeneration, and highlight inflammasome inhibition as a promising therapeutic avenue currently under investigation in preclinical and early clinical studies.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":"29-42"},"PeriodicalIF":13.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145574977","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lactation, tissue-resident immunity, and protection against breast cancer. 哺乳、组织常驻免疫和预防乳腺癌。
IF 13.9 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-01 Epub Date: 2025-12-31 DOI: 10.1016/j.it.2025.12.004
Martin Little, Samantha Ye, Benjamin P Fairfax

Parity and lactation have long been recognised as protective factors in breast cancer, with notable risk reduction in triple negative breast cancer (TNBC). Recent work by Virassamy et al. suggests a tissue-specific, persistent immune surveillance underpins this effect, particularly in women who have also breastfed.

胎次和哺乳期长期以来被认为是乳腺癌的保护因素,三阴性乳腺癌(TNBC)的风险显著降低。Virassamy等人最近的研究表明,一种组织特异性的、持续的免疫监测支持了这种效果,特别是在母乳喂养的女性中。
{"title":"Lactation, tissue-resident immunity, and protection against breast cancer.","authors":"Martin Little, Samantha Ye, Benjamin P Fairfax","doi":"10.1016/j.it.2025.12.004","DOIUrl":"10.1016/j.it.2025.12.004","url":null,"abstract":"<p><p>Parity and lactation have long been recognised as protective factors in breast cancer, with notable risk reduction in triple negative breast cancer (TNBC). Recent work by Virassamy et al. suggests a tissue-specific, persistent immune surveillance underpins this effect, particularly in women who have also breastfed.</p>","PeriodicalId":54412,"journal":{"name":"Trends in Immunology","volume":" ","pages":"3-5"},"PeriodicalIF":13.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145879455","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Trends in Immunology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
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