Lung DC-T immunity hub in immune surveillance: new concepts and future directions

IF 20.1 1区 医学 Q1 ONCOLOGY
Juan Liu, Boyi Cong, Xuetao Cao
{"title":"Lung DC-T immunity hub in immune surveillance: new concepts and future directions","authors":"Juan Liu,&nbsp;Boyi Cong,&nbsp;Xuetao Cao","doi":"10.1002/cac2.12643","DOIUrl":null,"url":null,"abstract":"<p>An effective coordination of immune and non-immune cells is essential for generating optimal regional immunity to combat tumorigenesis and infection at barrier tissues such as lung. Regional immune structures such as inducible bronchus-associated lymphoid tissue (iBALT) and tertiary lymphoid structure (TLS) play essential roles in modulating lung local immune responses. While the identification of iBALTs or TLS is generally dependent on conventional histology, it remains poorly understood how immune cells are spatiotemporally coordinated in the lung at single-cell resolution to effectively eliminate malignant cells and invading pathogens. Recently studies have revealed the presence of dendritic cell (DC)-T immunity hubs in human lung with close association with tumor immunotherapy response [<span>1</span>], antiviral immunity [<span>2</span>], and inflammation resolution [<span>3</span>]. The identification of DC-T immunity hubs spatiotemporally delineates the pulmonary multicellular networks at single-cell level in modulating antitumor and antiviral immune response, and will have profound implications for the diagnosis and treatment of lung cancer and infection.</p><p>The integration of single-cell technologies with high-resolution spatial imaging methods has been applied to reveal the spatial landscapes of lung tumor microenvironment (TME) in relevance to caner development, clinical outcome, and therapy responsiveness [<span>4</span>]. Multicellular immunity hubs of C-X-C motif chemokine ligand 13-positive (CXCL13<sup>+</sup>) T cells with interferon-stimulated gene (ISG)-expressing myeloid cells and malignant cells are detected in the luminal surface of human colorectal cancer [<span>5</span>]. The same group further revealed the existence of DC-T immunity hub in lung cancer [<span>1</span>]. This lung DC-T immunity hub is composed of activated CCR7<sup>+</sup> lysosomal-associated membrane protein 3-positive (LAMP3<sup>+</sup>) DCs (also termed as mature DCs enriched with regulatory molecules, mregDCs), stem-like transcription factor 7-positive (TCF7<sup>+</sup>) programmed cell death 1-positive (PD-1<sup>+</sup>) CD8<sup>+</sup> T cells, and C-C motif chemokine ligand 19-positive (CCL19<sup>+</sup>) fibroblasts, and strongly associate with beneficial outcome of PD-1 blockade therapy. Chemokine and adhesion pathways are essential for the stability and spatial organization of DC-T hub, in consistence with the report that activated leukocyte cell adhesion molecule, ligand of CD6 (ALCAM/CD166) stabilizes DC-CD8 T cell interactions at early tumor stages against immune evasion [<span>6</span>]. An intratumoral niche consisting of mregDCs, CXCL13<sup>+</sup>CD4<sup>+</sup> T helper cells and progenitor CD8<sup>+</sup> T cells is also present in hepatocellular carcinoma and associates with response to PD-1 blockade [<span>7</span>].</p><p>While combination of single-cell RNA sequencing (scRNA-seq) and spatial imaging methods has significantly facilitated the identification of cellular and molecular interactions in immunological niches at transcriptional levels, the multiplexed tissue imaging further allows characterization of spatial molecular interactions at protein levels. One study using multiplexed imaging, quantitative spatial analysis, and machine learning has mapped the landscapes of lung tumors in mice and human, and identified the networks of interacting lymphocytes (“lymphonets”) as a distinctive feature of the anti-cancer immune response. Such lymphnets contain TCF1<sup>+</sup>PD-1<sup>+</sup>CD8<sup>+</sup> T cell progenitors and gain cytotoxic CD8<sup>+</sup> T cell populations to enhance anti-tumor responses [<span>8</span>].</p><p>It should also be noted that immune cells can be remodeled by TME to promote tumor malignancy and metastasis [<span>9</span>]. One recent study using advanced multiplex imaging techniques discovered that mregDC recruit regulatory T cells (Tregs) form a mregDC-Treg cell niche around lymphatic vessels in the peripheral tumor stroma. This peri-lymphatic niche prevents antigen trafficking to the draining lymph nodes (dLNs), thus inhibits anti-tumor responses and promotes tumor progression [<span>10</span>]. Similarly, LAMP3<sup>+</sup> DC expressing indoleamine 2,3-dioxygenase 1 (IDO1) was shown to interact with exhausted CD8<sup>+</sup> T cells and CD4<sup>+</sup> Treg cells in cervical cancers (CC), and inhibiting IDO1 could enhance the efficacy of immune checkpoint blockade treatment in a mouse model of CC [<span>11</span>]. Further elucidating the mechanisms for education and reprograming of the anti-tumor versus pro-tumor niche in TME will reveal novel targets for cancer screening, diagnosis, and treatment.</p><p>Although multiple studies have analyzed the immunological mechanisms underlying lung infection such as that caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [<span>12</span>], few has been understood about the tissue immunological niches along the entire process of infection. In a recent study, Cong <i>et al.</i> [<span>2</span>] integrated spatial enhanced resolution omics-sequencing (Stereo-seq) and scRNA-seq analysis, and identified specific immunity hub containing three co-localized immune cell subset, <i>Cd160</i><sup>+</sup><i>Cd8</i><sup>+</sup> T cells, tumor necrosis factor receptor superfamily, member 4 (<i>Tnfrsf4</i>)<i><sup>+</sup>Cd4<sup>+</sup></i> T cells and C-C motif chemokine receptor 7 (<i>Ccr7</i>)<i><sup>+</sup>Ido1<sup>+</sup></i> DCs which dynamically shapes host immunity along the entire process of SARS-CoV-2 infection (Figure 1). Chemokines, co-stimulatory factors and adhesion molecules are critical for the intercellular communication among hub components, emphasizing the active chemotaxis and adhesive responses of DC-T hub after viral infection.</p><p>The DC-T hub localizes in the alveoli, and provides the first critical defense against the invading microbial in the alveolar region, which is distinguished from iBALTs or gut-associated lymphoid tissues (GALTs) which only reside at deeper tissues of barrier organs [<span>2</span>] (Supplementary Table S1). The rapid response of DC-T immunity hub to SARS-CoV-2 infection as early as day 2 provides first-line defense against viral infection, challenging the traditional idea that T cell-mediated adaptive immunity requires 5∼7 days to take in place post infection. In addition, the rapid proliferation and potent interaction between <i>Cd160</i><sup>+</sup><i>Cd8</i><sup>+</sup> T cells and SLAM family member 9 (<i>Slamf9</i>)<sup>+</sup> macrophage is important for the clearance of SARS-CoV-2. The virally infected <i>Slamf9</i><sup>+</sup> macrophages highly express tissue remodeling and angiogenesis genes, implying their involvement in inflammation resolution and tissue remodeling.</p><p>As late as 14 days post infection, <i>Slamf9</i><sup>+</sup> macrophages differentiate toward triggering receptor expressed on myeloid cells 2 (<i>Trem2</i>)<sup>+</sup> and fructose-bisphosphatase 1 (<i>Fbp1</i>)<sup>+</sup> alveolar macrophages, accompanied by downregulation of inflammatory genes such as tumor necrosis factor (<i>Tnf</i>), complement component 1, q subcomponent (<i>C1q</i>), but restoration of inflammation resolution and tissue repair genes such as macrophage receptor with collagenous structure (<i>Marco</i>) and <i>Cd36</i>, emphasizing the importance of macrophage compartments of DC-T immunity hub in inflammation resolution and tissue repair after SARS-CoV-2 infection [<span>3</span>]. Moreover, <i>Slamf9</i><sup>+</sup> macrophages can interact with distinct neutrophil subpopulations via platelet and endothelial cell adhesion molecule (PECAM), CCL, CD80, and interleukin 10 (IL-10) pathways, contributing to their roles in inflammation resolution and tissue repair (Figure 2). In another study, an immune-epithelial progenitor niche is shown to restrain lung regeneration and drivespost-acute sequelae of corona virus disease 2019 (COVID-19) (PASC), further suggesting important and diverse roles of immunological niche in determining inflammation outcomes and clinal consequences post lung infections [<span>13</span>].</p><p>Immune metabolic mechanisms play essential roles in modulating immune cell function and behavior in tumor, infection and inflammation, and offer novel opportunities for prevention and treatment of related disorders. The <i>Ccr7<sup>+</sup>Ido1<sup>+</sup></i> DCs residing at the center of DC-T hub is phenotypically resembling mregDCs [<span>14</span>], and multiple evidence have identified the metabolic cues in control of the identity and fate of mregDCs. Glycolysis is activated upon CCR7 ligation in DCs and supports CCR7-medited DC migration via maintaining cytoskeleton rearrangement and receptor oligomerization, and thereby supporting tissue inflammation [<span>15</span>]. An intermediate metabolite of mevalonate pathway, farnesyl pyrophosphate (FPP), could enhance mregDC migration to dLNs via remodeling mitochondrial structure and metabolism, and consequently lead to sustained germinal center responses and pathological immune responses [<span>16</span>]. In addition, IDO1 expression in CCR7<sup>+</sup> DCs promotes the tolerogenic function of conventional DC2 (cDC2) via producing tryptophan metabolite l-kynurenine, suggesting an indispensable role for tryptophan metabolism in controlling the tolerogenic property of cDCs [<span>17</span>].</p><p>Metabolic crosstalk emerges as critical mechanisms for regulating DC-centered immunity during tumor and infection. Intra-tumoral glutamine supplementation could support cDC1-mediated CD8<sup>+</sup> T cell immunity to overcome therapeutic resistance to immunotherapies [<span>18</span>]. In addition, melanoma-derived lactate serves as a trigger for sterol regulatory element binding transcription factor 2 (SREBP2)-dependent activation of cholesterol metabolism in mregDCs within TME, forming a lactate-SREBP2 signaling axis driving mregDCs tolerogenic maturation and suppression of antitumor immunity. DC-specific ablation or inhibition of SREBP2 exert anti-tumor therapeutic effects by promoting antitumor CD8<sup>+</sup> T cell activation [<span>19</span>]. Moreover, hyperglycaemia inhibit antiviral adaptive immune response via shifting the composition and gene expression of distinct lung DC subsets, most notably cDC1. The increased glucose-to-acetyl-CoA metabolism induced by hyperglycaemia alters global chromatin and key function genes in DCs, suggesting an indispensable role of metabolic-immune pathway in orchestrating DC dysregulation in pulmonary viral infection [<span>20</span>]. It will be intriguing to identify whether glucose and cholesterol metabolism would affect the function of <i>Ccr7<sup>+</sup>Ido1<sup>+</sup></i> DC and its communications with T cells and B cells in the immunity hub during pulmonary cancer and infection.</p><p>In sum, the lung DC-T immunity hubs exhibit a unique multicellular network of CCR7-expressing DCs and distinct T cell subsets, orchestrating lung immunity during infection and cancer. These immunity hubs represent the previously unrecognized mechanisms for the dynamic intercellular communications to establish host immune surveillance and homeostasis. The key issues of biological function, regulation mechanism, and disease relevance of immunity hub remain largely unanswered and are worthy of further investigations in the future, for examples, (1) the distinct developmental origin, functional specialization and metabolic remodeling of the hub cellular components; (2) the regulatory mechanisms governing the initial formation, expansion, and restoration of the immunity hub; (3) the tissue antigens and niche signals that determine the functional polarization and migratory property of hub CCR7<sup>+</sup> DCs; (4) the distribution and characteristic of DC-T immunity hub at other barrier tissues such as gastrointestinal tract, etc. Future exploration of the comprehensive spatiotemporal lung immune landscape will have profound implications in understanding how regional immunity dictates the development of cancer and infection, and will greatly facilitate the development of effective immunotherapies based on key cells or molecules involved in DC-T immunity hub. The integrative multi-omics analysis techniques will also initiate a paradigm-shifting transformation to oncological and immunological studies and provide a comprehensive resource for the scientific community to understand lung diseases and developing therapies in future.</p><p>Xuetao Cao and Juan Liu conceived and conceptualized the concept of this writing and wrote the original draft. Boyi Cong generated the figures and table with supervision from Xuetao Cao and Juan Liu. All authors critically revised the manuscript.</p><p>The authors declare no conflicts of interest.</p><p>This work was supported by Grants from the National Key R&amp;D Program of China (2023YFA1801400) and National Natural Science Foundation of China (92374115 and 82388201).</p><p>Not applicable.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"45 3","pages":"209-214"},"PeriodicalIF":20.1000,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12643","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12643","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

An effective coordination of immune and non-immune cells is essential for generating optimal regional immunity to combat tumorigenesis and infection at barrier tissues such as lung. Regional immune structures such as inducible bronchus-associated lymphoid tissue (iBALT) and tertiary lymphoid structure (TLS) play essential roles in modulating lung local immune responses. While the identification of iBALTs or TLS is generally dependent on conventional histology, it remains poorly understood how immune cells are spatiotemporally coordinated in the lung at single-cell resolution to effectively eliminate malignant cells and invading pathogens. Recently studies have revealed the presence of dendritic cell (DC)-T immunity hubs in human lung with close association with tumor immunotherapy response [1], antiviral immunity [2], and inflammation resolution [3]. The identification of DC-T immunity hubs spatiotemporally delineates the pulmonary multicellular networks at single-cell level in modulating antitumor and antiviral immune response, and will have profound implications for the diagnosis and treatment of lung cancer and infection.

The integration of single-cell technologies with high-resolution spatial imaging methods has been applied to reveal the spatial landscapes of lung tumor microenvironment (TME) in relevance to caner development, clinical outcome, and therapy responsiveness [4]. Multicellular immunity hubs of C-X-C motif chemokine ligand 13-positive (CXCL13+) T cells with interferon-stimulated gene (ISG)-expressing myeloid cells and malignant cells are detected in the luminal surface of human colorectal cancer [5]. The same group further revealed the existence of DC-T immunity hub in lung cancer [1]. This lung DC-T immunity hub is composed of activated CCR7+ lysosomal-associated membrane protein 3-positive (LAMP3+) DCs (also termed as mature DCs enriched with regulatory molecules, mregDCs), stem-like transcription factor 7-positive (TCF7+) programmed cell death 1-positive (PD-1+) CD8+ T cells, and C-C motif chemokine ligand 19-positive (CCL19+) fibroblasts, and strongly associate with beneficial outcome of PD-1 blockade therapy. Chemokine and adhesion pathways are essential for the stability and spatial organization of DC-T hub, in consistence with the report that activated leukocyte cell adhesion molecule, ligand of CD6 (ALCAM/CD166) stabilizes DC-CD8 T cell interactions at early tumor stages against immune evasion [6]. An intratumoral niche consisting of mregDCs, CXCL13+CD4+ T helper cells and progenitor CD8+ T cells is also present in hepatocellular carcinoma and associates with response to PD-1 blockade [7].

While combination of single-cell RNA sequencing (scRNA-seq) and spatial imaging methods has significantly facilitated the identification of cellular and molecular interactions in immunological niches at transcriptional levels, the multiplexed tissue imaging further allows characterization of spatial molecular interactions at protein levels. One study using multiplexed imaging, quantitative spatial analysis, and machine learning has mapped the landscapes of lung tumors in mice and human, and identified the networks of interacting lymphocytes (“lymphonets”) as a distinctive feature of the anti-cancer immune response. Such lymphnets contain TCF1+PD-1+CD8+ T cell progenitors and gain cytotoxic CD8+ T cell populations to enhance anti-tumor responses [8].

It should also be noted that immune cells can be remodeled by TME to promote tumor malignancy and metastasis [9]. One recent study using advanced multiplex imaging techniques discovered that mregDC recruit regulatory T cells (Tregs) form a mregDC-Treg cell niche around lymphatic vessels in the peripheral tumor stroma. This peri-lymphatic niche prevents antigen trafficking to the draining lymph nodes (dLNs), thus inhibits anti-tumor responses and promotes tumor progression [10]. Similarly, LAMP3+ DC expressing indoleamine 2,3-dioxygenase 1 (IDO1) was shown to interact with exhausted CD8+ T cells and CD4+ Treg cells in cervical cancers (CC), and inhibiting IDO1 could enhance the efficacy of immune checkpoint blockade treatment in a mouse model of CC [11]. Further elucidating the mechanisms for education and reprograming of the anti-tumor versus pro-tumor niche in TME will reveal novel targets for cancer screening, diagnosis, and treatment.

Although multiple studies have analyzed the immunological mechanisms underlying lung infection such as that caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [12], few has been understood about the tissue immunological niches along the entire process of infection. In a recent study, Cong et al. [2] integrated spatial enhanced resolution omics-sequencing (Stereo-seq) and scRNA-seq analysis, and identified specific immunity hub containing three co-localized immune cell subset, Cd160+Cd8+ T cells, tumor necrosis factor receptor superfamily, member 4 (Tnfrsf4)+Cd4+ T cells and C-C motif chemokine receptor 7 (Ccr7)+Ido1+ DCs which dynamically shapes host immunity along the entire process of SARS-CoV-2 infection (Figure 1). Chemokines, co-stimulatory factors and adhesion molecules are critical for the intercellular communication among hub components, emphasizing the active chemotaxis and adhesive responses of DC-T hub after viral infection.

The DC-T hub localizes in the alveoli, and provides the first critical defense against the invading microbial in the alveolar region, which is distinguished from iBALTs or gut-associated lymphoid tissues (GALTs) which only reside at deeper tissues of barrier organs [2] (Supplementary Table S1). The rapid response of DC-T immunity hub to SARS-CoV-2 infection as early as day 2 provides first-line defense against viral infection, challenging the traditional idea that T cell-mediated adaptive immunity requires 5∼7 days to take in place post infection. In addition, the rapid proliferation and potent interaction between Cd160+Cd8+ T cells and SLAM family member 9 (Slamf9)+ macrophage is important for the clearance of SARS-CoV-2. The virally infected Slamf9+ macrophages highly express tissue remodeling and angiogenesis genes, implying their involvement in inflammation resolution and tissue remodeling.

As late as 14 days post infection, Slamf9+ macrophages differentiate toward triggering receptor expressed on myeloid cells 2 (Trem2)+ and fructose-bisphosphatase 1 (Fbp1)+ alveolar macrophages, accompanied by downregulation of inflammatory genes such as tumor necrosis factor (Tnf), complement component 1, q subcomponent (C1q), but restoration of inflammation resolution and tissue repair genes such as macrophage receptor with collagenous structure (Marco) and Cd36, emphasizing the importance of macrophage compartments of DC-T immunity hub in inflammation resolution and tissue repair after SARS-CoV-2 infection [3]. Moreover, Slamf9+ macrophages can interact with distinct neutrophil subpopulations via platelet and endothelial cell adhesion molecule (PECAM), CCL, CD80, and interleukin 10 (IL-10) pathways, contributing to their roles in inflammation resolution and tissue repair (Figure 2). In another study, an immune-epithelial progenitor niche is shown to restrain lung regeneration and drivespost-acute sequelae of corona virus disease 2019 (COVID-19) (PASC), further suggesting important and diverse roles of immunological niche in determining inflammation outcomes and clinal consequences post lung infections [13].

Immune metabolic mechanisms play essential roles in modulating immune cell function and behavior in tumor, infection and inflammation, and offer novel opportunities for prevention and treatment of related disorders. The Ccr7+Ido1+ DCs residing at the center of DC-T hub is phenotypically resembling mregDCs [14], and multiple evidence have identified the metabolic cues in control of the identity and fate of mregDCs. Glycolysis is activated upon CCR7 ligation in DCs and supports CCR7-medited DC migration via maintaining cytoskeleton rearrangement and receptor oligomerization, and thereby supporting tissue inflammation [15]. An intermediate metabolite of mevalonate pathway, farnesyl pyrophosphate (FPP), could enhance mregDC migration to dLNs via remodeling mitochondrial structure and metabolism, and consequently lead to sustained germinal center responses and pathological immune responses [16]. In addition, IDO1 expression in CCR7+ DCs promotes the tolerogenic function of conventional DC2 (cDC2) via producing tryptophan metabolite l-kynurenine, suggesting an indispensable role for tryptophan metabolism in controlling the tolerogenic property of cDCs [17].

Metabolic crosstalk emerges as critical mechanisms for regulating DC-centered immunity during tumor and infection. Intra-tumoral glutamine supplementation could support cDC1-mediated CD8+ T cell immunity to overcome therapeutic resistance to immunotherapies [18]. In addition, melanoma-derived lactate serves as a trigger for sterol regulatory element binding transcription factor 2 (SREBP2)-dependent activation of cholesterol metabolism in mregDCs within TME, forming a lactate-SREBP2 signaling axis driving mregDCs tolerogenic maturation and suppression of antitumor immunity. DC-specific ablation or inhibition of SREBP2 exert anti-tumor therapeutic effects by promoting antitumor CD8+ T cell activation [19]. Moreover, hyperglycaemia inhibit antiviral adaptive immune response via shifting the composition and gene expression of distinct lung DC subsets, most notably cDC1. The increased glucose-to-acetyl-CoA metabolism induced by hyperglycaemia alters global chromatin and key function genes in DCs, suggesting an indispensable role of metabolic-immune pathway in orchestrating DC dysregulation in pulmonary viral infection [20]. It will be intriguing to identify whether glucose and cholesterol metabolism would affect the function of Ccr7+Ido1+ DC and its communications with T cells and B cells in the immunity hub during pulmonary cancer and infection.

In sum, the lung DC-T immunity hubs exhibit a unique multicellular network of CCR7-expressing DCs and distinct T cell subsets, orchestrating lung immunity during infection and cancer. These immunity hubs represent the previously unrecognized mechanisms for the dynamic intercellular communications to establish host immune surveillance and homeostasis. The key issues of biological function, regulation mechanism, and disease relevance of immunity hub remain largely unanswered and are worthy of further investigations in the future, for examples, (1) the distinct developmental origin, functional specialization and metabolic remodeling of the hub cellular components; (2) the regulatory mechanisms governing the initial formation, expansion, and restoration of the immunity hub; (3) the tissue antigens and niche signals that determine the functional polarization and migratory property of hub CCR7+ DCs; (4) the distribution and characteristic of DC-T immunity hub at other barrier tissues such as gastrointestinal tract, etc. Future exploration of the comprehensive spatiotemporal lung immune landscape will have profound implications in understanding how regional immunity dictates the development of cancer and infection, and will greatly facilitate the development of effective immunotherapies based on key cells or molecules involved in DC-T immunity hub. The integrative multi-omics analysis techniques will also initiate a paradigm-shifting transformation to oncological and immunological studies and provide a comprehensive resource for the scientific community to understand lung diseases and developing therapies in future.

Xuetao Cao and Juan Liu conceived and conceptualized the concept of this writing and wrote the original draft. Boyi Cong generated the figures and table with supervision from Xuetao Cao and Juan Liu. All authors critically revised the manuscript.

The authors declare no conflicts of interest.

This work was supported by Grants from the National Key R&D Program of China (2023YFA1801400) and National Natural Science Foundation of China (92374115 and 82388201).

Not applicable.

Abstract Image

肺DC-T免疫中枢在免疫监测中的应用:新概念及未来发展方向。
免疫细胞和非免疫细胞的有效协调对于产生最佳的区域免疫以对抗肿瘤发生和肺等屏障组织的感染至关重要。诱导支气管相关淋巴组织(iBALT)和三级淋巴组织(TLS)等区域免疫结构在调节肺局部免疫应答中发挥重要作用。虽然iBALTs或TLS的鉴定通常依赖于传统组织学,但人们对免疫细胞如何在单细胞分辨率下在肺中进行时空协调以有效消除恶性细胞和入侵的病原体仍然知之甚少。最近的研究表明,树突状细胞(DC)-T免疫中枢在人肺中存在,与肿瘤免疫治疗反应[1]、抗病毒免疫[2]和炎症消退[3]密切相关。DC-T免疫枢纽的发现在时空上揭示了单细胞水平上肺多细胞网络在调节抗肿瘤和抗病毒免疫反应中的作用,并将对肺癌和感染的诊断和治疗产生深远的影响。单细胞技术与高分辨率空间成像方法的结合已被应用于揭示与癌症发展、临床结果和治疗反应性相关的肺肿瘤微环境(TME)的空间景观。C-X-C基元趋化因子配体13阳性(CXCL13+) T细胞表达干扰素刺激基因(ISG)骨髓细胞和恶性细胞的多细胞免疫中枢在人类结直肠癌[5]的管腔表面被检测到。同一组进一步揭示了DC-T免疫中枢在肺癌[1]中的存在。这种肺DC-T免疫中枢由活化的CCR7+溶酶体相关膜蛋白3阳性(LAMP3+) dc(也称为富含调控分子的成熟dc, mregdc)、干细胞样转录因子7阳性(TCF7+)程序性细胞死亡1阳性(PD-1+) CD8+ T细胞和C-C基序趋化因子配体19阳性(CCL19+)成纤维细胞组成,并与PD-1阻断治疗的有益结果密切相关。趋化因子和粘附途径对于DC-T hub的稳定性和空间组织至关重要,与活化的白细胞粘附分子CD6配体(ALCAM/CD166)在肿瘤早期稳定DC-CD8 T细胞相互作用以抵抗免疫逃逸[6]的报道一致。由mregdc、CXCL13+CD4+ T辅助细胞和祖细胞CD8+ T细胞组成的肿瘤内生态位也存在于肝细胞癌中,并与PD-1阻断剂[7]的应答有关。单细胞RNA测序(scRNA-seq)和空间成像方法的结合极大地促进了在转录水平上免疫龛中细胞和分子相互作用的识别,而多重组织成像进一步允许在蛋白质水平上表征空间分子相互作用。一项使用多路复用成像、定量空间分析和机器学习的研究绘制了小鼠和人类肺肿瘤的版图,并确定了相互作用淋巴细胞(“淋巴细胞”)的网络是抗癌免疫反应的一个显著特征。这些淋巴网含有TCF1+PD-1+CD8+ T细胞祖细胞,并获得细胞毒性CD8+ T细胞群,从而增强抗肿瘤反应[8]。还应注意的是,TME可以重塑免疫细胞,促进肿瘤的恶性和转移。最近的一项研究使用先进的多重成像技术发现,mregDC募集调节性T细胞(Tregs)在周围肿瘤基质的淋巴管周围形成mregDC- treg细胞生态位。这个淋巴周围生态位阻止抗原运输到引流淋巴结(dln),从而抑制抗肿瘤反应并促进肿瘤进展[10]。同样,表达吲哚胺2,3-双加氧酶1 (IDO1)的LAMP3+ DC在宫颈癌(CC)中与耗尽的CD8+ T细胞和CD4+ Treg细胞相互作用,抑制IDO1可以增强CC[11]小鼠模型中免疫检查点阻断治疗的效果。进一步阐明TME中抗肿瘤与促肿瘤生态位的教育和重编程机制,将为癌症筛查、诊断和治疗提供新的靶点。虽然已有多项研究分析了SARS-CoV-2等肺部感染的免疫学机制,但对感染全过程的组织免疫生态位了解甚少。在最近的一项研究中,Cong等人。 [2]整合了空间增强分辨率组学测序(Stereo-seq)和scRNA-seq分析,发现了包含三个共定位免疫细胞亚群的特异性免疫枢纽,Cd160+Cd8+ T细胞,肿瘤坏死因子受体超家族,成员4 (Tnfrsf4)+Cd4+ T细胞和C-C基序趋化因子受体7 (Ccr7)+Ido1+ dc,在整个SARS-CoV-2感染过程中动态塑造宿主免疫(图1)。共刺激因子和粘附分子是中枢成分细胞间通讯的关键,强调DC-T中枢在病毒感染后的活性趋化性和粘附反应。DC-T中枢位于肺泡内,为抵御肺泡区微生物入侵提供了第一道关键防御,这与仅存在于屏障器官[2]深层组织的ibalt或肠相关淋巴组织(galt)不同(补充表S1)。DC-T免疫中心对SARS-CoV-2感染的快速反应最早在第2天就提供了针对病毒感染的第一线防御,挑战了T细胞介导的适应性免疫在感染后需要5 ~ 7天才能发生的传统观点。此外,Cd160+Cd8+ T细胞与SLAM家族成员9 (Slamf9)+巨噬细胞的快速增殖和强有力的相互作用对于清除SARS-CoV-2很重要。病毒感染的Slamf9+巨噬细胞高度表达组织重塑和血管生成基因,这意味着它们参与炎症消退和组织重塑。在感染后14天,Slamf9+巨噬细胞分化为表达于髓样细胞2 (Trem2)+和果糖双磷酸酶1 (Fbp1)+肺泡巨噬细胞的触发受体,伴随着肿瘤坏死因子(Tnf)、补体成分1、q亚成分(C1q)等炎症基因的下调,但炎症消退和组织修复基因如胶原结构巨噬细胞受体(Marco)和Cd36的恢复。强调DC-T免疫中枢巨噬细胞区室在SARS-CoV-2感染后炎症消退和组织修复中的重要性。此外,Slamf9+巨噬细胞可以通过血小板和内皮细胞粘附分子(PECAM)、CCL、CD80和白细胞介素10 (IL-10)途径与不同的中性粒细胞亚群相互作用,有助于它们在炎症消退和组织修复中的作用(图2)。在另一项研究中,免疫上皮祖细胞生态位被证明可以抑制肺再生并驱动2019冠状病毒病(COVID-19) (PASC)的急性后后遗症。进一步提示免疫生态位在决定肺部感染后炎症结局和临床后果方面的重要和多样化作用。免疫代谢机制在调节肿瘤、感染和炎症的免疫细胞功能和行为中发挥着重要作用,为相关疾病的预防和治疗提供了新的机会。位于DC-T枢纽中心的Ccr7+Ido1+ dc在表型上类似于mregdc[14],多种证据已经确定了控制mregdc身份和命运的代谢线索。糖酵解在DC中被CCR7连接激活,并通过维持细胞骨架重排和受体寡聚化来支持CCR7介导的DC迁移,从而支持组织炎症[15]。甲羟丙酸途径的中间代谢物法尼酯焦磷酸盐(FPP)可以通过重塑线粒体结构和代谢来促进mregDC向dln的迁移,从而导致持续的生发中心反应和病理免疫反应[16]。此外,IDO1在CCR7+ DCs中的表达通过产生色氨酸代谢物l-犬尿氨酸来促进常规DC2 (cDC2)的耐受性功能,提示色氨酸代谢在控制cDCs的耐受性方面发挥着不可或缺的作用。代谢串扰是肿瘤和感染期间调节dc中心免疫的关键机制。肿瘤内补充谷氨酰胺可支持cdc1介导的CD8+ T细胞免疫克服对免疫疗法的治疗性耐药[18]。此外,黑色素瘤来源的乳酸可触发TME内mregDCs中胆固醇代谢的固醇调节元件结合转录因子2 (SREBP2)依赖性激活,形成乳酸-SREBP2信号轴,驱动mregDCs耐受性成熟和抗肿瘤免疫抑制。dc特异性消融或抑制SREBP2通过促进抗肿瘤CD8+ T细胞活化[19]发挥抗肿瘤治疗作用。此外,高血糖通过改变不同肺DC亚群的组成和基因表达来抑制抗病毒适应性免疫反应,尤其是cDC1。 高血糖诱导的葡萄糖-乙酰辅酶a代谢增加改变了DC的整体染色质和关键功能基因,表明代谢-免疫途径在肺部病毒感染[20]中协调DC失调中发挥了不可或缺的作用。在肺癌和感染期间,葡萄糖和胆固醇代谢是否会影响Ccr7+Ido1+ DC的功能及其与免疫中枢T细胞和B细胞的通讯,这将是一个有趣的研究。总之,肺DC-T免疫中枢表现出一个独特的多细胞网络,由表达ccr7的dc和不同的T细胞亚群组成,在感染和癌症期间协调肺免疫。这些免疫中枢代表了以前未被认识的动态细胞间通讯机制,以建立宿主免疫监视和稳态。免疫中枢的生物学功能、调控机制和疾病相关性等关键问题仍有待进一步研究,如:(1)中枢细胞组分的独特发育起源、功能特化和代谢重塑;(2)免疫中枢初始形成、扩展和恢复的调控机制;(3)决定中枢CCR7+ DCs功能极化和迁移特性的组织抗原和生态位信号;(4) DC-T免疫中枢在胃肠道等其他屏障组织的分布及特点。未来对肺部综合时空免疫景观的探索将对理解区域免疫如何决定癌症和感染的发展具有深远的意义,并将极大地促进基于DC-T免疫中枢参与的关键细胞或分子的有效免疫疗法的开发。综合多组学分析技术也将启动范式转换到肿瘤学和免疫学研究,并为科学界未来了解肺部疾病和开发治疗方法提供全面的资源。曹雪涛和刘娟构思并构思了这个写作的概念,并撰写了初稿。丛伯义在曹雪涛和刘娟的监督下生成了图表。所有作者都严格修改了手稿。作者声明无利益冲突。国家重点发展计划项目(2023YFA1801400)和国家自然科学基金项目(92374115和82388201)资助。不适用。
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 求助全文
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信