1
|
Bosma DMT, Busselaar J, Staal MD, Frijlink E, Mack M, Salerno F, Borst J. CD4 + T-cell help delivery to monocyte-derived dendritic cells promotes effector differentiation of helper and cytotoxic T cells. Immunol Lett 2025; 275:107022. [PMID: 40239818 DOI: 10.1016/j.imlet.2025.107022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Delivery of CD4+ T-cell help optimizes CD8+ T-cell effector and memory responses via CD40-mediated licensing of conventional dendritic cells (DCs). Using comparative vaccination settings that prime CD8+ T cells in presence or absence of CD4+ T-cell help, we observed that CD4+ T-cell activation promoted influx of monocytes into the vaccine-draining lymph nodes (dLNs), where they differentiated into monocyte-derived (Mo)DCs, as defined by the most recent standards. Abrogation of these responses by CCR2-targeted depletion indicated that monocyte-derived cells in the dLN promoted T-helper 1 (Th1) type effector differentiation of CD4+ T cells, as well as effector differentiation of CD8+ T cells. Monocyte-derived cells in dLNs upregulated CD40, CD80 and PD-L1 as a result of CD4+ T-cell help. The response of monocyte-derived cells to CD4+ T-cell help was independent of natural killer (NK) cells and proceeded via CD40 ligand (L)-CD40 interactions and IFNγ signaling. Our data argue for a scenario wherein activated CD4+ T cells in dLNs crosstalk via CD40L and IFNγ signals to monocytes, promoting their local differentiation into MoDCs. This event enhances formation of CD4+ Th1 and CD8+ cytotoxic effector T cell pool, most likely by virtue of their improved costimulatory status and cytokine production.
Collapse
Affiliation(s)
- Douwe M T Bosma
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Julia Busselaar
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mo D Staal
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Elselien Frijlink
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Fiamma Salerno
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jannie Borst
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
2
|
Calindi A, Ehrlich LIR. Intrathymic Regulation of Dendritic Cell Subsets and Their Contributions to Central Tolerance. Immunol Rev 2025; 332:e70039. [PMID: 40433811 PMCID: PMC12117523 DOI: 10.1111/imr.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/27/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
Thymic dendritic cells (DCs) are critical mediators of central tolerance, cooperating with medullary thymic epithelial cells (mTECs) and B cells to establish T-cell self-tolerance to the proteome. The DC compartment is highly heterogeneous and is comprised of three major subsets, plasmacytoid dendritic cells (pDCs) and two conventional dendritic cell (cDC) subsets, cDC1 and cDC2. Thymic cDC1 and cDC2 arise from distinct progenitors and access the thymus at different stages of their differentiation, but both become activated by cellular and secreted cues received within the sterile thymus environment. Activated cDC1s and cDC2s have been implicated in presenting distinct types of self-antigens to induce central tolerance. Thus, understanding how the distinct cDC subsets are regulated within the thymus environment will provide important insights into mechanisms governing self-tolerance. Furthermore, the thymic DC compartment undergoes age-associated compositional and transcriptional changes that likely impact the efficiency and quality of central tolerance established over the lifespan. Here, we review recent findings from our lab and others on mechanisms regulating thymic DC activation, the distinct roles of thymic DC subsets in central tolerance, and age-associated changes in thymic DCs that could impact T-cell selection.
Collapse
Affiliation(s)
- Aparna Calindi
- Department of Molecular BiosciencesThe University of Texas at AustinAustinTexasUSA
| | - Lauren I. R. Ehrlich
- Department of Molecular BiosciencesThe University of Texas at AustinAustinTexasUSA
- LaMontagne Center for Infectious DiseaseThe University of Texas at AustinAustinTexasUSA
| |
Collapse
|
3
|
Aliazis K, Christofides A, Shah R, Yeo YY, Jiang S, Charest A, Boussiotis VA. The tumor microenvironment's role in the response to immune checkpoint blockade. NATURE CANCER 2025:10.1038/s43018-025-00986-3. [PMID: 40514448 DOI: 10.1038/s43018-025-00986-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 04/17/2025] [Indexed: 06/16/2025]
Abstract
Beyond cancer cells, the tumor microenvironment (TME) includes cells of the innate and adaptive immune systems but also non-immune cells, such as fibroblasts and endothelial cells. Depending on the cues they receive, infiltrating myeloid cells, such as monocytes, macrophages, dendritic cells and neutrophils, perform immune stimulatory or suppressive functions by educating adaptive immune cells, thereby guiding their responses to cancer cells and cancer treatment, such as immune checkpoint blockade (ICB). The increasing understanding that anti-tumor immunity goes beyond T cells with improved functionality, and the unraveling of resistance mechanisms beyond T cell exhaustion, have renewed interest in non-T cell components of the TME to identify novel therapeutic targets and improve ICB responses. Here, we review immune and non-immune cellular components of the TME that regulate adaptive cell responses and their role in ICB response and resistance.
Collapse
Affiliation(s)
- Konstantinos Aliazis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Anthos Christofides
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Rushil Shah
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ohio State University Medical College, Columbus, OH, USA
| | - Yao Yu Yeo
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sizun Jiang
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alain Charest
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vassiliki A Boussiotis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Yuan Z, Shu L, Zheng Y, Wang Y, Zheng M, Sun J, Fu J, Zhou Z, Song S, Liu Z, Li F, Cai Z. IRF8 Drives Conventional Type 1 Dendritic Cell Differentiation and CD8 + T Cell Activation to Aggravate Abdominal Aortic Aneurysm Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416238. [PMID: 40184622 PMCID: PMC12165085 DOI: 10.1002/advs.202416238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/04/2025] [Indexed: 04/06/2025]
Abstract
Abdominal aortic aneurysm (AAA) is the most common true aneurysm worldwide, and recent studies suggest that dendritic cells (DCs) play a key role in its development, though the specific subtypes and underlying mechanisms remain unclear. In this study, the role of interferon regulatory factor 8 (IRF8) in AAA is investigated by focusing on its effect on the differentiation of DC precursors into conventional type 1 dendritic cells (cDC1s). It is found significant infiltration of HLA-DR+ IRF8+ cells in human AAA tissue samples. In mice, DC-specific overexpression of Irf8 exacerbates aneurysm expansion following periadventitial elastase application, while DC-specific Irf8 deletion attenuates AAA development. Batf3-/- mice, which lack cDC1s, exhibit AAA characteristics similar to the Irf8-deleted mice. Additionally, an increased population of activated CD8+ T cells is observed in the DC-Irf8 overexpressed mice, while the DC-Irf8 deletion mice show a decrease in these cells. Blocking antigen cross-presentation to CD8+ T cells also reduces AAA progression. Tissue microarray analysis of human aortic samples further confirms a correlation between IRF8 expression and AAA development. These findings suggest that IRF8 activation promotes cDC1 differentiation, leading to the recruitment of CD8+ T cells, which contribute to aortic wall destruction and AAA formation.
Collapse
Affiliation(s)
- Zhen Yuan
- Department of CardiologyThe Second Affiliated Hospital, School of MedicineHeart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- State Key Laboratory of Transvascular Implantation DevicesHangzhou310009China
- Heart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- Transvascular Implantation Devices Research InstituteHangzhou310053China
| | - Li Shu
- Department of CardiologyThe Second Affiliated Hospital, School of MedicineHeart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- State Key Laboratory of Transvascular Implantation DevicesHangzhou310009China
- Heart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- Transvascular Implantation Devices Research InstituteHangzhou310053China
| | - Yidan Zheng
- Department of Cardiovascular SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yidong Wang
- Department of CardiologyThe Second Affiliated Hospital, School of MedicineHeart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- State Key Laboratory of Transvascular Implantation DevicesHangzhou310009China
- Heart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- Transvascular Implantation Devices Research InstituteHangzhou310053China
| | - Mengsha Zheng
- Department of PathologyThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Jie Sun
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel DiseasesBeijing100029China
| | - Jiantao Fu
- Clinical Center for HIV/AIDSBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
| | - Zihao Zhou
- Department of Cardiovascular SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Shen Song
- State Key Laboratory of Cardiovascular DiseaseFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Zhenjie Liu
- Department of Vascular SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Fei Li
- Department of Cardiovascular SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Structural Heart DiseaseFuwai Yunnan Cardiovascular HospitalKunming650102China
| | - Zhejun Cai
- Department of CardiologyThe Second Affiliated Hospital, School of MedicineHeart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- State Key Laboratory of Transvascular Implantation DevicesHangzhou310009China
- Heart Regeneration and Repair Key Laboratory of Zhejiang ProvinceHangzhou310009China
- Transvascular Implantation Devices Research InstituteHangzhou310053China
| |
Collapse
|
5
|
Hushmandi K, Imani Fooladi AA, Reiter RJ, Farahani N, Liang L, Aref AR, Nabavi N, Alimohammadi M, Liu L, Sethi G. Next-generation immunotherapeutic approaches for blood cancers: Exploring the efficacy of CAR-T and cancer vaccines. Exp Hematol Oncol 2025; 14:75. [PMID: 40382583 DOI: 10.1186/s40164-025-00662-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/25/2025] [Indexed: 05/20/2025] Open
Abstract
Recent advancements in immunotherapy, particularly Chimeric antigen receptor (CAR)-T cell therapy and cancer vaccines, have significantly transformed the treatment landscape for leukemia. CAR-T cell therapy, initially promising in hematologic cancers, faces notable obstacles in solid tumors due to the complex and immunosuppressive tumor microenvironment. Challenges include the heterogeneous immune profiles of tumors, variability in antigen expression, difficulties in therapeutic delivery, T cell exhaustion, and reduced cytotoxic activity at the tumor site. Additionally, the physical barriers within tumors and the immunological camouflage used by cancer cells further complicate treatment efficacy. To overcome these hurdles, ongoing research explores the synergistic potential of combining CAR-T cell therapy with cancer vaccines and other therapeutic strategies such as checkpoint inhibitors and cytokine therapy. This review describes the various immunotherapeutic approaches targeting leukemia, emphasizing the roles and interplay of cancer vaccines and CAR-T cell therapy. In addition, by discussing how these therapies individually and collectively contribute to tumor regression, this article aims to highlight innovative treatment paradigms that could enhance clinical outcomes for leukemia patients. This integrative approach promises to pave the way for more effective and durable treatment strategies in the oncology field. These combined immunotherapeutic strategies hold great promise for achieving more complete and lasting remissions in leukemia patients. Future research should prioritize optimizing treatment sequencing, personalizing therapeutic combinations based on individual patient and tumor characteristics, and developing novel strategies to enhance T cell persistence and function within the tumor microenvironment. Ultimately, these efforts will advance the development of more effective and less toxic immunotherapeutic interventions, offering new hope for patients battling this challenging disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | | | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
6
|
Chen J, Liu TT, Ou F, Ohara RA, Jo S, Postoak JL, Egawa T, Day RB, Murphy TL, Murphy KM, Kim S. C/EBPα activates Irf8 expression in myeloid progenitors at the +56-kb enhancer to initiate cDC1 development. Sci Immunol 2025; 10:eadt5899. [PMID: 40378239 DOI: 10.1126/sciimmunol.adt5899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/18/2025] [Indexed: 05/18/2025]
Abstract
Development of type 1 conventional dendritic cells (cDC1s) underlies the capacity to generate antiviral and antitumor immune responses. Here, we identify the basis for cDC1 development from its earliest progenitors, determining the hierarchy of several required transcription factors and uncovering a series of mandatory cis interactions between constituent enhancers within the Irf8 superenhancer. We produced in vivo mutations of two C/EBPα binding sites that comprise the Irf8 +56-kilobase (kb) enhancer that markedly reduced IRF8 expression in all myeloid progenitors and impaired cDC1 development. These sites did not bind RUNX1 or RUNX3, and C/EBPα expression was instead regulated by their action at the Cebpa +37-kb enhancer, placing RUNX factors upstream of Cebpa in regulating Irf8. Last, we demonstrate that cis interactions between the +56-kb Irf8 enhancer and the previously reported +41- and +32-kb Irf8 enhancers are mandatory in the sequential progression of these stage-specific constituent elements.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Feiya Ou
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ray A Ohara
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Suin Jo
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Joshua Luke Postoak
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Takeshi Egawa
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ryan B Day
- Section of Stem Cell Biology, Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sunkyung Kim
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Microbiology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
7
|
Zhou J, Tison K, Zhou H, Bai L, Acharyya RK, McEachern D, Metwally H, Wang Y, Pitter M, Choi JE, Vatan L, Liao P, Yu J, Lin H, Jiang L, Wei S, Gao X, Grove S, Parolia A, Cieslik M, Kryczek I, Green MD, Lin JX, Chinnaiyan AM, Leonard WJ, Wang S, Zou W. STAT5 and STAT3 balance shapes dendritic cell function and tumour immunity. Nature 2025:10.1038/s41586-025-09000-3. [PMID: 40369063 DOI: 10.1038/s41586-025-09000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Immune checkpoint blockade (ICB) has transformed cancer therapy1,2. The efficacy of immunotherapy depends on dendritic cell-mediated tumour antigen presentation, T cell priming and activation3,4. However, the relationship between the key transcription factors in dendritic cells and ICB efficacy remains unknown. Here we found that ICB reprograms the interplay between the STAT3 and STAT5 transcriptional pathways in dendritic cells, thereby activating T cell immunity and enabling ICB efficacy. Mechanistically, STAT3 restrained the JAK2 and STAT5 transcriptional pathway, determining the fate of dendritic cell function. As STAT3 is often activated in the tumour microenvironment5, we developed two distinct PROTAC (proteolysis-targeting chimera) degraders of STAT3, SD-36 and SD-2301. STAT3 degraders effectively degraded STAT3 in dendritic cells and reprogrammed the dendritic cell-transcriptional network towards immunogenicity. Furthermore, STAT3 degrader monotherapy was efficacious in treatment of advanced tumours and ICB-resistant tumours without toxicity in mice. Thus, the crosstalk between STAT3 and STAT5 transcriptional pathways determines the dendritic cell phenotype in the tumour microenvironment and STAT3 degraders hold promise for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiajia Zhou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Kole Tison
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Haibin Zhou
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Longchuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ranjan Kumar Acharyya
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Donna McEachern
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hoda Metwally
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yu Wang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Pitter
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jae Eun Choi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Long Jiang
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xue Gao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Michael D Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shaomeng Wang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA.
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA.
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Liu Z, Jiang X, Ke Z, Wang W, Tang J, Dai Y. PAR2 deficiency impairs antitumor immunity and attenuates anti-PD1 efficacy in colorectal cancer. Pharmacol Res 2025; 215:107721. [PMID: 40174816 DOI: 10.1016/j.phrs.2025.107721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
A T cell-inflamed tumor microenvironment is predictive of better prognosis and clinical response to immunotherapy. Proteinase-activated receptor 2 (PAR2), a member of G-protein coupled receptors is involved in inflammatory process and the progression of various cancers. However, the role of PAR2 in modulating the tumor microenvironment remains unclear. Here, we found that PAR2 high-expression was associated with a favorable prognosis in patients with colorectal cancer. Intriguingly, PAR2 expression in human colorectal cancer was mainly confined to tumor cells and was significantly associated with CD8+ T cell infiltration. Tumor-intrinsic PAR2 deficiency blunted antitumor immune responses to promote tumor growth and attenuated the therapeutic efficacy of anti-PD1 in a mouse model of colon cancer. Tumors with downregulated PAR2 showed decreased CD8+ T cell infiltration and impaired effector function. Mechanistically, PAR2 activation in tumor cells induced CXCL9 and CXCL10 production via PI3K/AKT/mTOR signaling, thereby enhancing CD8+ T cell recruitment in the tumor microenvironment. In addition, PAR2 was essential for dendritic cell activation and differentiation towards conventional type 1 subset. PAR2 deficiency in dendritic cells markedly impaired their ability to prime CD8+ T cells and control tumor growth in vivo. Thus, our findings identify new roles for PAR2 in promoting antitumor immunity and provide a promising target to improve immunotherapy efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Zilin Liu
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Xuehui Jiang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Weihong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jianqiang Tang
- Department of General Surgery, Peking University First Hospital, Beijing, China.
| | - Yun Dai
- Department of Gastroenterology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
9
|
Tao M, Liu W, Chen J, Liu R, Zou J, Yu B, Wang C, Huang M, Chen Q, Zhang Z, Chen Z, Sun H, Zhou C, Tan S, Zheng Y, Wang H. Transcriptome Landscape of Cancer-Associated Fibroblasts in Human PDAC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415196. [PMID: 40019403 PMCID: PMC12120754 DOI: 10.1002/advs.202415196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/27/2025] [Indexed: 03/01/2025]
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in the progression of pancreatic ductal adenocarcinoma (PDAC). Here, integrated single-cell RNA sequencing analysis is utilized to comprehensively map CAFs in the human PDAC tumor microenvironment (TME). Normal fibroblasts (NFs) and nine distinct CAF subtypes are identified including newly identified CAF subtypes, CDCP1+FTL+ CAFs, transitional CAFs (tCAFs), interferon simulated genes (ISG)+ myofibroblastic CAFs (myCAFs), and proliferative CAFs (pCAFs). CDCP1+FTL+ CAFs, pCAFs, and ISG+ myCAFs are associated with unfavorable clinical outcomes. CDCP1+FTL+ CAFs exhibit enhanced glycolysis and iron metabolism, resisting ferroptosis. The antigen-presenting CAFs (apCAFs) show high heterogeneity, consisting of multiple subtypes expressing distinct immune cell signatures. The CAF subtypes display differentiation plasticity, transitioning from early normal-like CAFs (nCAFs) to inflammatory CAFs (iCAFs) and myCAFs, ultimately leading to more invasive pCAFs. AP-1 family members FOS and JUN regulate the malignant phenotype conversion of NFs to nCAFs, while transforming growth factor-β (TGFβ) and interferon-γ (IFNγ) signals trigger the interconversion between classic myCAFs and iCAFs, respectively. A close interaction between CAFs and myeloid cells (especially neutrophils) is further observed in PDAC-TME, mainly mediated by CXCR4-CXCL12 chemotaxis. This work depicts a detailed CAF map and its dynamic interconvertible shift, providing important insights for combined targeted CAFs therapy.
Collapse
Affiliation(s)
- Mengyu Tao
- Department of OncologyShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200800P. R. China
| | - Wenting Liu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Jianhua Chen
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Rujiao Liu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Jianling Zou
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Bo Yu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Chenchen Wang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Mingzhu Huang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Qingjian Chen
- Department of OncologyShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200800P. R. China
| | - Zhe Zhang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Zhiyu Chen
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Haoyu Sun
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Department of ImmunologySchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Cheng Zhou
- Department of Radiation OncologyNanfang HospitalSouthern Medical UniversityGuangzhou510515P. R. China
| | - Shuguang Tan
- The Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Yuxuan Zheng
- Human Phenome InstituteMinhang HosptialFudan UniversityShanghai201203P. R. China
| | - Hongxia Wang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| |
Collapse
|
10
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
11
|
Ludwig AL, McKay ZP, Carter GP, Katz MA, Howell G, Jain V, Arvai S, Dittmer DP, Bigner DD, Ashley DM, Shoaf ML, Desjardins A, Gregory SG, Brown MC, Gromeier M. Lymphotropic Virotherapy Engages DC and High Endothelial Venule Inflammation to Mediate Cancer In Situ Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.23.25326279. [PMID: 40313264 PMCID: PMC12045412 DOI: 10.1101/2025.04.23.25326279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Intratumor (IT) inoculation of the rhino:poliovirus chimera, PVSRIPO, yielded objective radiographic responses with long-term survival in 20% of patients with recurrent glioblastoma (rGBM). PVSRIPO infects dendritic cells (DCs) and sets up non-cytopathogenic viral (v)RNA replication, which triggers sustained type-I IFN (IFN-I) signaling and antitumor T cell priming. Here we identify IFN-I signaling in glioma-draining cervical lymph nodes (cLN) as a mediator of polio virotherapy. Transient IFN-I signaling after IT therapy was rescued by cervical perilymphatic injection (CPLI) of PVSRIPO, targeting cLN directly. Dual-site (IT+CPLI) PVSRIPO induced profound inflammatory reprogramming of cLN, enhanced vRNA replication and IFN-I signaling in DCs and High Endothelial Venules (HEV), augmented anti-glioma efficacy in mice, and was associated with T cell activation in rGBM patients. A Ph2 clinical trial of IT+CPLI PVSRIPO is ongoing ( NCT06177964 ). This work implicates the lymphatic system as a novel virotherapy target and demonstrates the CPLI concept to complement brain tumor immunotherapy.
Collapse
|
12
|
Barger LN, Wang D, Saravia A, Mezzano V, Ward G, Loomis C, Feldman C, Tuluc M, Seedor RS, Gaskill PJ, Coghill AE, Suneja G, Dehzangi I, Hope JL, Jour G, Romano G. Population analysis and immunologic landscape of melanoma in people living with HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.17.648995. [PMID: 40313919 PMCID: PMC12045344 DOI: 10.1101/2025.04.17.648995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
People living with HIV (PLWH) diagnosed with melanoma have consistently exhibited worse clinical outcomes than HIV-negative individuals (PLw/oH) with the same cancer, even in the era of antiretroviral therapy (ART). To investigate the underlying factors contributing to these disparities, we analyzed electronic health records from 922 PLWH and 334,972 PLw/oH with melanoma. PLWH were diagnosed with melanoma at a younger age and had a higher representation of Hispanic and Black individuals. Notably, PLWH had a markedly increased risk of brain metastases. Additionally, despite similar treatment durations, PLWH experienced significant delays in initiating immune checkpoint therapy (ICI) and exhibited worse survival outcomes at both five- and ten-years post-treatment with ICI. To explore potential biological determinants of these disparities, we conducted spatial immune transcriptomics on melanoma tumors (n=11). This analysis revealed a more immunosuppressive tumor landscape in PLWH, characterized by upregulated immune checkpoints (e.g., PD1, LAG3, CTLA4) and diminished antigen presentation (e.g., HLA-DRB, B2M ), with distinct spatial distributions in the tumors versus the tumor microenvironments. Downstream validation via multiplex immunofluorescence (n=15 PLWH, n=14 PLw/oH) confirmed an exhausted CD8 + T cell compartment, marked by enrichment of PD1 int LAG3 - and PD1 int LAG3 + subpopulations, along with a significant accumulation of immunosuppressive myeloid-derived suppressor cells (CD11b + HLA-DR - CD33 + ) in PLWH. These distinct immune profiles suggest chronic HIV infection fosters a permissive tumor microenvironment that might undermine effective immune responses and contribute to poor clinical outcomes for PLWH with melanoma. Targeting the actionable immune pathways identified in this study could inform tailored therapeutic strategies to mitigate these disparities.
Collapse
|
13
|
Luo Z, Li X, Zhu D, Fu W, Liu Y, Zheng L, Chen P, Gong C, Liu X. Implantable Immunostimulant Microneedle Patch for Post-Surgical Prevention of Cancer Recurrence and Distant Tumor Inhibition. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22362-22374. [PMID: 40194999 DOI: 10.1021/acsami.5c01155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cancer recurrence after surgical resection remains a grand challenge in achieving long-term eradication. Here, we develop a biocompatible and implantable immunostimulant microneedle patch designed to suppress local tumor recurrence after surgery. The patch, fabricated using methacrylate-modified hyaluronic acid, incorporates 2'3'-cGAMP, a STING agonist, and IL-2, a cytokine approved for clinical cancer immunotherapy that expands T cells. The patch enables controlled release of cGAMP to induce dendritic cell maturation, antitumor macrophage polarization (M1 macrophage), and T cell priming and activation. Simultaneously, localized IL-2 activates CD8+ T cells and recruits immune cells to the tumor microenvironment. When combined with an anti-CTLA-4 antibody, an immune checkpoint blockade, the hybrid microneedle patch significantly reduces Treg cells at the surgery sites, enhancing immune responses and effectively inhibiting the progression of distant tumors in both prophylactic and therapeutic models. Compared with traditional postsurgical chemotherapy and radiotherapy, this patch-mediated immunotherapy demonstrates superior efficacy in mitigating tumor relapse while offering higher biocompatibility. Our findings suggest that this immunotherapeutic patch has potential as a translational tool to prevent cancer recurrence in patients with resectable tumors.
Collapse
Affiliation(s)
- Zichao Luo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031 China
- NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031 China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200031 China
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
| | - Xinchao Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dandan Zhu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
| | - Wangxian Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxia Liu
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
| | - Lewen Zheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
- Skin Research Institute of Singapore, 308232 Singapore
- Lee Kong Chian School of Medicine, Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921 Singapore
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaogang Liu
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 138634, Singapore
| |
Collapse
|
14
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
15
|
Lisicka W, Earley ZM, Sifakis JJ, Erickson SA, Mattingly JR, Wu-Woods NJ, Krishnamurthy SR, Belkaid Y, Ismagilov RF, Cyster JG, Riesenfeld SJ, Bendelac A, Jabri B. Immunoglobulin A controls intestinal virus colonization to preserve immune homeostasis. Cell Host Microbe 2025; 33:498-511.e10. [PMID: 40154490 DOI: 10.1016/j.chom.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/26/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Immunoglobulin A (IgA) is the predominant immunoglobulin isotype in mammals, primarily secreted at type I mucosal surfaces. Despite its abundance, the precise role of secretory IgA in the intestinal lumen, where it coats a diverse array of commensal microbiota, has remained elusive. Our study reveals that germinal center IgA responses are essential for preventing chronic colonization of the gut by specific viruses. In the absence of IgA, chronic viral colonization triggers an antigen-driven expansion of CD8αβ+ intraepithelial lymphocytes (IELs). Although these IELs are unable to clear the virus, they contribute to maintaining homeostasis by regulating its load and type I interferon responses. Consequently, IgA deficiency increases susceptibility to colitis in genetically susceptible hosts or following chemical induction but only in the presence of viral pathobionts requiring IgA for their clearance. These findings underscore the potential vulnerability of IgA-deficient individuals to immunopathology when exposed to selective viral pathobionts.
Collapse
Affiliation(s)
- Wioletta Lisicka
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Zachary M Earley
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph J Sifakis
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Steven A Erickson
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Jonathan R Mattingly
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Natalie J Wu-Woods
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Siddharth R Krishnamurthy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rustem F Ismagilov
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Samantha J Riesenfeld
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA; Paris City University, Imagine Institute, Paris, France.
| |
Collapse
|
16
|
Song X, Zhou Z, Liu J, Li J, Yu C, Zeh HJ, Klionsky DJ, Stockwell BR, Wang J, Kang R, Kroemer G, Tang D. Cytosolic cytochrome c represses ferroptosis. Cell Metab 2025:S1550-4131(25)00149-4. [PMID: 40233758 DOI: 10.1016/j.cmet.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/08/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
The release of cytochrome c, somatic (CYCS) from mitochondria to the cytosol is an established trigger of caspase-dependent apoptosis. Here, we unveil an unexpected role for cytosolic CYCS in inhibiting ferroptosis-a form of oxidative cell death driven by uncontrolled lipid peroxidation. Mass spectrometry and site-directed mutagenesis revealed the existence of a cytosolic complex composed of inositol polyphosphate-4-phosphatase type I A (INPP4A) and CYCS. This CYCS-INPP4A complex is distinct from the CYCS-apoptotic peptidase activating factor 1 (APAF1)-caspase-9 apoptosome formed during mitochondrial apoptosis. CYCS boosts INPP4A activity, leading to increased formation of phosphatidylinositol-3-phosphate, which prevents phospholipid peroxidation and plasma membrane rupture, thus averting ferroptotic cell death. Unbiased screening led to the identification of the small-molecule compound 10A3, which disrupts the CYCS-INPP4A interaction. 10A3 sensitized cultured cells and tumors implanted in immunocompetent mice to ferroptosis. Collectively, these findings redefine our understanding of cytosolic CYCS complexes that govern diverse cell death pathways.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhuan Zhou
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Department of Critical Care Medicine, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Jingbo Li
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
17
|
Wang J, Gong P, Liu Q, Wang M, Wu D, Li M, Zheng S, Wang H, Long Q. Stimulation of regulatory dendritic cells suppresses cytotoxic T cell function and alleviates DEN-induced liver injury, fibrosis and hepatocellular carcinoma. Front Immunol 2025; 16:1565486. [PMID: 40264769 PMCID: PMC12011597 DOI: 10.3389/fimmu.2025.1565486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Background Dendritic cells (DCs) are versatile professional antigen-presenting cells and play an instrumental role in the generation of antigen-specific T-cell responses. Modulation of DC function holds promise as an effective strategy to improve anti-tumor immunotherapy efficacy and enhance self-antigen tolerance in autoimmune diseases. Methods Wild-type (WT) and TLR2 knockout (KO) mice at 2 weeks of age were injected intraperitoneally (i.p.) with a single dose of diethylnitrosamine (DEN) to induce hepatocellular carcinoma (HCC). Four weeks later, WT and KO mice were randomly divided into control and treatment groups and treated once every two days for 30 weeks with phosphate buffered saline (PBS) and a mix of 4 TLR2-activating lactic acid-producing probiotics (LAP), respectively. Mice were euthanized after 30 weeks of LAP treatment and their liver tissues were collected for gene expression, histological, flow cytometric and single-cell RNA sequencing analyses. Results We demonstrate here that oral administration of a mix of TLR2-activating LAP triggers a marked accumulation of regulatory DCs (rDCs) in the liver of mice. LAP-treated mice are protected from DEN-induced liver injury, fibrosis and HCC in a TLR2-dependent manner. Single-cell transcriptome profiling revealed that LAP treatment determines an immunosuppressive hepatic T-cell program that is characterized by a significantly reduced cytotoxic activity. The observed functional changes of T cells correlated well with the presence of a hepatic DC subset displaying a regulatory or tolerogenic transcriptional signature. Conclusion Overall, these data suggest that stimulation of regulatory dendritic cells (rDCs) in the liver by LAP suppresses cytotoxic T-cell function and alleviates DEN-induced liver damage, fibrosis and tumorigenesis.
Collapse
Affiliation(s)
- Junjie Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Pixu Gong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qingqing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Menglei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Dengfang Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Mengyu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Shujie Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qiaoming Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
18
|
Mauvais FX, Hamel Y, Silvin A, Mulder K, Hildner K, Akyol R, Dalod M, Koumantou D, Saveanu L, Garfa M, Cagnard N, Bertocci B, Ginhoux F, van Endert P. Metallophilic marginal zone macrophages cross-prime CD8 + T cell-mediated protective immunity against blood-borne tumors. Immunity 2025; 58:843-860.e20. [PMID: 40139188 DOI: 10.1016/j.immuni.2025.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/27/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
Splenic metallophilic marginal zone macrophages (MMMs) are positioned to control the dissemination of blood-borne threats. We developed a purification protocol to enable characterization of MMMs phenotypically and transcriptionally. MMM gene expression profile was enriched for pathways associated with CD8+ T cell activation and major histocompatibility complex class I (MHC class I) cross-presentation. In vitro, purified MMMs equaled conventional dendritic cells type 1 (cDC1s) in cross-priming CD8+ T cells to soluble and particulate antigens, yet MMMs employed a distinct vacuolar processing pathway. In vivo biphoton and ex vivo light-sheet imaging showed long-standing contacts with cognate T cells differentiating to effectors. MMMs cross-primed protective CD8+ T cell antitumor responses both by capturing blood-borne tumor antigens and by internalizing tumor cells seeding the spleen. This cross-priming required expression of the transcription factor Batf3 by MMMs but was independent of cDC1-mediated capture of tumor material for cross-presentation or MHC class I-dressing. Thus, MMMs combine control of the dissemination of blood-borne pathogens and tumor materials with the initiation of innate and adaptive responses.
Collapse
Affiliation(s)
- François-Xavier Mauvais
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Service de Physiologie - Explorations Fonctionnelles Pédiatriques, AP-HP, Hôpital Universitaire Robert Debré, 75019 Paris, France.
| | - Yamina Hamel
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France
| | - Aymeric Silvin
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kai Hildner
- University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Medical Department 1, Deutsches Zentrum Immuntherapie, 91054 Erlangen, Germany
| | - Ramazan Akyol
- Aix Marseille Université, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Dalod
- Aix Marseille Université, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Despoina Koumantou
- Université Paris Cité, Centre de recherche sur l'inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018 Paris, France; Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018 Paris, France
| | - Loredana Saveanu
- Université Paris Cité, Centre de recherche sur l'inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018 Paris, France; Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018 Paris, France
| | - Meriem Garfa
- Cell Imaging, Structure Fédérative de Recherche Necker, INSERM, US24/CNRS UMS3633, 75015 Paris, France
| | - Nicolas Cagnard
- Bioinformatics Core Facilities, Structure Fédérative de Recherche Necker, INSERM, US24/CNRS UMS3633, 75015 Paris, France
| | - Barbara Bertocci
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France; Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, (A∗STAR), Singapore, Singapore; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Peter van Endert
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 75015 Paris, France.
| |
Collapse
|
19
|
Jo S, Ohara RA, Theisen DJ, Kim S, Liu T, Bullock CB, He M, Ou F, Chen J, Piersma SJ, Postoak JL, Yokoyama WM, Diamond MS, Murphy TL, Murphy KM. Shared pathway of WDFY4-dependent cross-presentation of immune complexes by cDC1 and cDC2. J Exp Med 2025; 222:e20240955. [PMID: 39918736 PMCID: PMC11804880 DOI: 10.1084/jem.20240955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/18/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Priming CD8+ T cells against tumors or viral pathogens results largely from cross-presentation of exogenous antigens by type 1 conventional dendritic cells (cDC1s). Although monocyte-derived DCs and cDC2s can cross-present in vitro, their physiological relevance remains unclear. Here, we used genetic models to evaluate the role of cDC subsets in presentation of cell-associated and immune complex antigens to CD4+ and CD8+ T cells in vivo. For cell-associated antigens, cDC1s were necessary and sufficient to prime both CD4+ and CD8+ T cells. In contrast, for immune complex antigens, either cDC1 or cDC2, but not monocyte-derived DCs, could carry out cross-presentation to CD8+ T cells. Mice lacking cDC1 and vaccinated with immune complexes could cross-prime CD8+ T cells that were sufficient to mediate tumor rejection. Notably, this cross-presentation mediated by cDC2 was also WDFY4 dependent, similar to cross-presentation of cell-associated antigens by cDC1. These results demonstrate a previously unrecognized activity of WDFY4 in cDC2s and suggest a cross-presentation pathway shared by cDC subsets.
Collapse
Affiliation(s)
- Suin Jo
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ray A. Ohara
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Derek J. Theisen
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sunkyung Kim
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Tiantian Liu
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Christopher B. Bullock
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Michelle He
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Feiya Ou
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jing Chen
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sytse J. Piersma
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - J. Luke Postoak
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Wayne M. Yokoyama
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Theresa L. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
20
|
Martinez RJ, Hogquist KA. Sterile production of interferons in the thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf048. [PMID: 40184034 DOI: 10.1093/jimmun/vkaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
T-cell central tolerance is controlled by thymocyte TCR recognition of self-peptides presented by thymic APCs. While thymic epithelial cells are essential for T-cell central tolerance, a variety of other traditional APCs also play critical roles in T-cell selection. Similar to how peripheral APCs require activation to become effective, thymic APCs also require activation to become tolerogenic. Recent studies have identified IFNs as an essential factor for the activation and generation of an optimally tolerogenic thymic environment. In this review, we focus on interferon (IFN) production within the thymus and its effects on thymic APCs and developing thymocytes. We also examine the importance of T-cell tolerance to IFN itself as well as to interferon-stimulated proteins generated during peripheral immune responses.
Collapse
Affiliation(s)
- Ryan J Martinez
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
21
|
Luri-Rey C, Teijeira Á, Wculek SK, de Andrea C, Herrero C, Lopez-Janeiro A, Rodríguez-Ruiz ME, Heras I, Aggelakopoulou M, Berraondo P, Sancho D, Melero I. Cross-priming in cancer immunology and immunotherapy. Nat Rev Cancer 2025; 25:249-273. [PMID: 39881005 DOI: 10.1038/s41568-024-00785-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/31/2025]
Abstract
Cytotoxic T cell immune responses against cancer crucially depend on the ability of a subtype of professional antigen-presenting cells termed conventional type 1 dendritic cells (cDC1s) to cross-present antigens. Cross-presentation comprises redirection of exogenous antigens taken from other cells to the major histocompatibility complex class I antigen-presenting machinery. In addition, once activated and having sensed viral moieties or T helper cell cooperation via CD40-CD40L interactions, cDC1s provide key co-stimulatory ligands and cytokines to mount and sustain CD8+ T cell immune responses. This regulated process of cognate T cell activation is termed cross-priming. In cancer mouse models, CD8+ T cell cross-priming by cDC1s is crucial for the efficacy of most, if not all, immunotherapy strategies. In patients with cancer, the presence and abundance of cDC1s in the tumour microenvironment is markedly associated with the level of T cell infiltration and responsiveness to immune checkpoint inhibitors. Therapeutic strategies to increase the numbers of cDC1s using FMS-like tyrosine kinase 3 ligand (FLT3L) and/or their activation status show evidence of efficacy in cancer mouse models and are currently being tested in initial clinical trials with promising results so far.
Collapse
Affiliation(s)
- Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Álvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Stefanie K Wculek
- Innate Immune Biology Laboratory, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Carlos de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Claudia Herrero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | - Ignacio Heras
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Departments of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
22
|
Watts TH, Yeung KKM, Yu T, Lee S, Eshraghisamani R. TNF/TNFR Superfamily Members in Costimulation of T Cell Responses-Revisited. Annu Rev Immunol 2025; 43:113-142. [PMID: 39745933 DOI: 10.1146/annurev-immunol-082423-040557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Prosurvival tumor necrosis factor receptor (TNFR) superfamily (TNFRSF) members on T cells, including 4-1BB, CD27, GITR, and OX40, support T cell accumulation during clonal expansion, contributing to T cell memory. During viral infection, tumor necrosis factor superfamily (TNFSF) members on inflammatory monocyte-derived antigen-presenting cells (APCs) provide a postpriming signal (signal 4) for T cell accumulation, particularly in the tissues. Patients with loss-of-function mutations in TNFR/TNFSF members reveal a critical role for 4-1BB and CD27 in CD8 T cell control of Epstein-Barr virus and other childhood infections and of OX40 in CD4 T cell responses. Here, on the 20th anniversary of a previous Annual Review of Immunology article about TNFRSF signaling in T cells, we discuss the effects of endogenous TNFRSF signals in T cells upon recognition of TNFSF members on APCs; the role of TNFRSF members, including TNFR2, on regulatory T cells; and recent advances in the incorporation of TNFRSF signaling in T cells into immunotherapeutic strategies for cancer.
Collapse
Affiliation(s)
- Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | - Karen K M Yeung
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | - Tianning Yu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | - Seungwoo Lee
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | | |
Collapse
|
23
|
Catena X, Contreras-Alcalde M, Juan-Larrea N, Cerezo-Wallis D, Calvo TG, Mucientes C, Olmeda D, Suárez J, Oterino-Sogo S, Martínez L, Megías D, Sancho D, Tejedo C, Frago S, Dudziak D, Seretis A, Stoitzner P, Soengas MS. Systemic rewiring of dendritic cells by melanoma-secreted midkine impairs immune surveillance and response to immune checkpoint blockade. NATURE CANCER 2025; 6:682-701. [PMID: 40155713 DOI: 10.1038/s43018-025-00929-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 02/11/2025] [Indexed: 04/01/2025]
Abstract
Cutaneous melanomas express a high number of potential neoepitopes, yet a substantial fraction of melanomas shift into immunologically cold phenotypes. Using cellular systems, mouse models and large datasets, we identify the tumor-secreted growth factor midkine (MDK) as a multilayered inhibitor of antigen-presenting cells. Mechanistically, MDK acts systemically in primary tumors, lymph nodes and the bone marrow, promoting a STAT3-mediated impairment of differentiation, activation and function of dendritic cells (DCs), particularly, conventional type 1 DCs (cDC1s). Furthermore, MDK rewires DCs toward a tolerogenic state, impairing CD8+ T cell activation. Downregulating MDK improves DC-targeted vaccination, CD40 agonist treatment and immune checkpoint blockade in mouse models. Moreover, we present an MDK-associated signature in DCs that defines poor prognosis and immune checkpoint blockade resistance in individuals with cancer. An inverse correlation between MDK- and cDC1-associated signatures was observed in a variety of tumor types, broadening the therapeutic implications of MDK in immune-refractory malignancies.
Collapse
Affiliation(s)
- Xavier Catena
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Contreras-Alcalde
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Naiara Juan-Larrea
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Daniela Cerezo-Wallis
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Yale University School of Medicine, New Haven, CT, USA
| | - Tonantzin G Calvo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cynthia Mucientes
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Instituto de Investigaciones Biomédicas Sols-Morreale, Madrid, Spain
| | - Javier Suárez
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sergio Oterino-Sogo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Lola Martínez
- Flow Cytometry Core Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Diego Megías
- Advanced Optical Microscopy Unit, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| | - David Sancho
- Immunobiology Lab, Centro Nacional de Investigación Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Tejedo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Susana Frago
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Diana Dudziak
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
- Comprehensive Cancer Center Central Germany Jena/Leipzig, Jena, Germany
| | - Athanasios Seretis
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|
24
|
Ahvati H, Roudi R, Sobhani N, Safari F. CD47 as a potent target in cancer immunotherapy: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189294. [PMID: 40057140 DOI: 10.1016/j.bbcan.2025.189294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Cancer is the second-highest cause of death worldwide. Accordingly, finding new cancer treatments is of great interest to researchers. The current platforms to fight cancer such as chemotherapy, radiotherapy, and surgery are limited in efficacy, especially in the metastatic setting. In this war against cancer, the immune system is a powerful ally, but tumor cells often outsmart it through alternative pathways. Cluster of differentiation 47 (CD47), a protein that normally prevents healthy cells from being attacked by immune cells, is often overexpressed on cancer cells. This makes CD47 a prime target for immunotherapy. Blocking of CD47 has the potential to unleash the immune system's cell populations-such as myeloid cells, macrophages, and T cells-to allow the immune system to discover and destroy cancer cells more successfully. In this review, we aimed to provide the latest information and findings about the roles of CD47 in the regulation of various cellular pathways and, thus, the importance of CD47 as a potential target in cancer therapy.
Collapse
Affiliation(s)
- Hiva Ahvati
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| |
Collapse
|
25
|
Hu MM, Zhao Y, Zhang N, Gong FY, Zhang W, Dong CS, Dai JF, Wang J. Tumor Microenvironment: Obstacles and Opportunities for T Cell-Based Tumor Immunotherapies. Mol Cancer Res 2025; 23:277-287. [PMID: 39898773 DOI: 10.1158/1541-7786.mcr-24-0747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/20/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The complex composition and dynamic change of the tumor microenvironment (TME), mainly consisting of tumor cells, immune cells, stromal cells, and extracellular components, significantly impede the effector function of cytotoxic T lymphocytes (CTL), thus representing a major obstacle for tumor immunotherapies. In this review, we summarize and discuss the impacts and underlying mechanisms of major elements in the TME (different cell types, extracellular matrix, nutrients and metabolites, etc.) on the infiltration, survival, and effector functions of T cells, mainly CD8+ CTLs. Moreover, we also highlight recent advances that may potentiate endogenous antitumor immunity and improve the efficacy of T cell-based immunotherapies in patients with cancer by manipulating components inside/outside of the TME. A deeper understanding of the effects and action mechanisms of TME components on the tumor-eradicating ability of CTLs may pave the way for discovering new targets to augment endogenous antitumor immunity and for designing combinational therapeutic regimens to enhance the efficacy of tumor immunotherapies in the clinic.
Collapse
Affiliation(s)
- Miao-Miao Hu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Nan Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Fang-Yuan Gong
- Department of Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wei Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chun-Sheng Dong
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jian-Feng Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
26
|
Lai YG, Liao HT, Chen YH, Huang SW, Liou YH, Wu ZQ, Liao NS. cGAS and STING in Host Myeloid Cells Are Essential for Effective Cyclophosphamide Treatment of Advanced Breast Cancer. Cancers (Basel) 2025; 17:1130. [PMID: 40227734 PMCID: PMC11987962 DOI: 10.3390/cancers17071130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cyclophosphamide (CTX) treatment in vivo kills proliferating tumor cells by DNA crosslinking; however, the suppression of tumor growth by CTX in several murine models requires CD8+ T cells. Given that CTX induces DNA damage and type I interferon (IFN-I), we investigated the role of host cGAS and STING in the anti-tumor effect of CTX in vivo. METHODS A metastasized EO771 breast cancer model with chromosomal instability and bone marrow (BM) chimera approach were used in this study. RESULTS We found that CTX therapy induces long-term survival of the mice, with this outcome being dependent on CD8+ T cells and cGAS/STING of BM-derived cells. Furthermore, the STING of type 1 conventional dendritic cells (cDC1s) and LysM+ cells and the IFN-I response of non-cDC1 myeloid cells are essential for CTX efficacy. We also found that the cGAS and STING of BM-derived cells positively modulate intratumoral exhausted and stem-cell-like CD8+ T cell populations under CTX treatment, with the latter only being affected by cGAS. CONCLUSIONS Our study demonstrates that the CD8+-T-cell-dependent anti-tumor mechanisms of CTX critically involve the cGAS-STING-IFN-I axis, IFN-I response, and STING-independent cGAS function in host myeloid cells. These findings suggest the deployment of CTX in treating advanced solid tumor to bypass the often-failed IFN-I production by tumor cells due to the chronic activation of intrinsic cGAS-STING caused by chromosomal instability.
Collapse
Affiliation(s)
- Yein-Gei Lai
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Hao-Ting Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
- Department of Life Sciences, National Central University, Taoyuan 320, Taiwan
| | - Yung-Hsiang Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Shih-Wen Huang
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Yae-Huei Liou
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Zhen-Qi Wu
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Nan-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| |
Collapse
|
27
|
Kumagai S, Momoi Y, Nishikawa H. Immunogenomic cancer evolution: A framework to understand cancer immunosuppression. Sci Immunol 2025; 10:eabo5570. [PMID: 40153489 DOI: 10.1126/sciimmunol.abo5570] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 06/26/2024] [Accepted: 03/05/2025] [Indexed: 03/30/2025]
Abstract
The process of tumor development involves tumor cells eluding detection and suppression of immune responses, which can cause decreased tumor cell antigenicity, expression of immunosuppressive molecules, and immunosuppressive cell recruitment to the tumor microenvironment (TME). Immunologically and genomically integrated analysis (immunogenomic analysis) of patient specimens has revealed that oncogenic aberrant signaling is involved in both carcinogenesis and immune evasion. In noninflamed cancers such as epidermal growth factor receptor (EGFR)-mutated lung cancers, genetic abnormalities in cancer cells contribute to the formation of an immunosuppressive TME by recruiting immunosuppressive cells, which cannot be fully explained by the cancer immunoediting hypothesis. This review summarizes the latest findings regarding the links between cancer genetic abnormalities and immunosuppression causing clinical resistance to immunotherapy. We propose the concepts of immunogenomic cancer evolution, in which cancer cell genomic evolution shapes the immunosuppressive TME, and immunogenomic precision medicine, in which cancer immunotherapy can be combined with molecularly targeted reagents that modulate the immunosuppressive TME.
Collapse
Affiliation(s)
- Shogo Kumagai
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Division of Cellular Signaling, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
| | - Yusaku Momoi
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Department of Tumor Pathology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
- Kindai University Faculty of Medicine, Osaka-sayama 589-8511, Japan
| |
Collapse
|
28
|
Rodriguez LIL, Amadio R, Piperno GM, Benvenuti F. Tissue-specific properties of type 1 dendritic cells in lung cancer: implications for immunotherapy. J Immunother Cancer 2025; 13:e010547. [PMID: 40132908 PMCID: PMC11938230 DOI: 10.1136/jitc-2024-010547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/22/2025] [Indexed: 03/27/2025] Open
Abstract
Checkpoint inhibitors have led to remarkable benefits in non-small cell lung cancer (NSCLC), yet response rates remain below expectations. High-dimensional analysis and mechanistic experiments in clinical samples and relevant NSCLC models uncovered the immune composition of lung cancer tissues, providing invaluable insights into the functional properties of tumor-infiltrating T cells and myeloid cells. Among myeloid cells, type 1 conventional dendritic cells (cDC1s) stand out for their unique ability to induce effector CD8 T cells against neoantigens and coordinate antitumoral immunity. Notably, lung resident cDC1 are particularly abundant and long-lived and express a unique tissue-specific gene program, underscoring their central role in lung immunity. Here, we discuss recent insights on the induction and regulation of antitumoral T cell responses in lung cancer, separating it from the tissue-agnostic knowledge generated from heterogeneous tumor models. We focus on the most recent studies dissecting functional states and spatial distribution of lung cDC1 across tumor stages and their impact on T cell responses to neoantigens. Finally, we highlight relevant gaps and emerging strategies to harness lung cDC1 immunostimulatory potential.
Collapse
Affiliation(s)
| | - Roberto Amadio
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Department of Life Sciences (DSV), University of Trieste, Trieste, Italy
| | - Giulia Maria Piperno
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Federica Benvenuti
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
29
|
Chakraborty AK, Kroehling L, Raut RD, Choudhury C, Kukuruzinska M, Gutkind JS, Varelas X, Sahay B, Monti S, Bais MV. LSD1 inhibition corrects dysregulated MHC-I and dendritic cells activation through IFNγ-CXCL9-CXCR3 axis to promote antitumor immunity in HNSCC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643710. [PMID: 40166238 PMCID: PMC11956948 DOI: 10.1101/2025.03.17.643710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Poor infiltration of CD8+ T cells and dysregulated MHC-I confer resistance to anticancer clinical therapies. This study aimed to elucidate the mechanisms of lysine-specific demethylase 1 (LSD1, encoded by KDM1A gene) in antitumor immunity in Head and Neck Squamous cell carcinoma (HNSCC). LSD1 inhibition in syngeneic and chronic tobacco carcinogen-induced HNSCC mice recruited activated dendritic cells (DCs), CD4+ and CD8+ T cells, enriched interferon-gamma (IFNγ) in T cells, CXCL9 in DCs, and CXCR3 in T cells, as evaluated using flow cytometry and single-cell RNA-seq analysis. Humanized HNSCC mice and TCGA data validated the inverse correlation of KDM1A with DC markers, CD8+ T cells, and their activating chemokines. Kdm1a knockout in mouse HNSCC and LSD1 inhibitor treatment to co-culture of human HNSCC cells with human peripheral blood mononuclear cells (PBMCs) resulted in MHC-I upregulation in cancer cells for efficient antigen presentation in tumors. Overall, LSD1 inhibition in tumor cells upregulates MHC class I and induces DCs to produce CXCL9, which in turn activates CD8+ T cells through the CXCL9-CXCR3 axis to produce IFNγ. Finally, we identified a novel mechanism by which LSD1 inhibition promotes the activation of H3K4me2 and its direct interaction with MHC-I to induce antitumor immunity. This may have implications in poorly immunogenic and immunotherapy-resistant cancers. Statement of Significance LSD1-mediated unique mechanisms have impact on epigenetic therapy, MHC-I resistant HNSCC therapies, and poor CD8+ and dendritic cell infilterated tumors.
Collapse
|
30
|
van der Geest R, Lee JS. Role of the basic leucine zipper transcription factor BATF2 in modulating immune responses and inflammation in health and disease. J Leukoc Biol 2025; 117:qiae245. [PMID: 39504573 PMCID: PMC11953073 DOI: 10.1093/jleuko/qiae245] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 11/08/2024] Open
Abstract
BATF2 is a transcription factor known to exhibit tumor-suppressive activity in cancer cells. Within recent years, however, BATF2 has also emerged as an important transcriptional regulator of the immune system. Through its immunomodulatory function, BATF2 has been implicated in a variety of (patho)physiological processes, including host defense against infection, antitumor immunity, and maintenance of tissue inflammatory homeostasis. Below, we discuss recent literature that has provided insight into the role of BATF2 as a transcriptional regulator of immune responses in health and disease, including the cell types that express BATF2, the different diseases in which the immunomodulatory effects of BATF2 have been shown to play a role, and the molecular mechanisms through which BATF2 is thought to exert those effects. In doing so, we highlight that the immunological effects of BATF2 are highly context dependent, and we point out the overlap between the mechanisms of action of BATF2 in infectious and noninfectious diseases. We also discuss areas of interest for future research, the clinical relevance of better understanding BATF2 function, and potential strategies for therapeutic modulation of BATF2.
Collapse
Affiliation(s)
- Rick van der Geest
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Janet S Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213, United States
- Vascular Medicine Institute, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, United States
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
| |
Collapse
|
31
|
Mujal AM, Owyong M, Santosa EK, Sauter JC, Grassmann S, Pedde AM, Meiser P, Wingert CK, Pujol M, Buchholz VR, Lau CM, Böttcher JP, Sun JC. Splenic TNF-α signaling potentiates the innate-to-adaptive transition of antiviral NK cells. Immunity 2025; 58:585-600.e6. [PMID: 40023159 DOI: 10.1016/j.immuni.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/29/2024] [Accepted: 02/07/2025] [Indexed: 03/04/2025]
Abstract
Natural killer (NK) cells possess both innate and adaptive features. Here, we investigated NK cell activation across tissues during cytomegalovirus infection, which generates antigen-specific clonal expansion and long-lived memory responses. Longitudinal tracking and single-cell RNA sequencing of NK cells following infection revealed enhanced activation in the spleen, as well as early formation of a CD69lo precursor population that preferentially gave rise to adaptive NK cells. Splenic NK cells demonstrated heightened tumor necrosis factor alpha (TNF-α) signaling and increased expression of the receptor TNFR2, which coincided with elevated TNF-α production by splenic myeloid cells. TNFR2-deficient NK cells exhibited impaired interferon gamma (IFN-γ) production and expansion. TNFR2 signaling engaged two distinct nuclear factor κB (NF-κB) signaling arms-innate effector NK cell responses required canonical NF-κB signaling, whereas non-canonical NF-κB signaling enforced differentiation of CD69lo adaptive NK cell precursors. Thus, NK cell priming in the spleen during viral infection promotes an innate-to-adaptive transition, providing insight into avenues for generating adaptive NK cell immunity across diverse settings.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Animals
- Mice
- Signal Transduction/immunology
- Spleen/immunology
- Immunity, Innate
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/immunology
- NF-kappa B/metabolism
- Adaptive Immunity
- Mice, Inbred C57BL
- Lymphocyte Activation/immunology
- Cytomegalovirus Infections/immunology
- Mice, Knockout
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Interferon-gamma/metabolism
- Muromegalovirus/immunology
- Antigens, Differentiation, T-Lymphocyte
- Antigens, CD
- Lectins, C-Type
Collapse
Affiliation(s)
- Adriana M Mujal
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Mark Owyong
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA
| | - Endi K Santosa
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA
| | - John C Sauter
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna-Marie Pedde
- Department of Experimental Immunology, Institute of Immunology, University of Tübingen, Tübingen, Germany; M3 Research Center, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; Institute of Molecular Immunology, TUM University Hospital, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Philippa Meiser
- Institute of Molecular Immunology, TUM University Hospital, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Claire K Wingert
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marine Pujol
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Colleen M Lau
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jan P Böttcher
- Department of Experimental Immunology, Institute of Immunology, University of Tübingen, Tübingen, Germany; M3 Research Center, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; Institute of Molecular Immunology, TUM University Hospital, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
32
|
Fan Y, Ji X, Yuan K, Wu Q, Lou M. HDAC1 Mediates Immunosuppression of the Tumor Microenvironment in Non-Small Cell Lung Cancer. J Inflamm Res 2025; 18:3333-3347. [PMID: 40078575 PMCID: PMC11900795 DOI: 10.2147/jir.s509316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Background Studies have demonstrated that histone deacetylase 1 (HDAC1) enables cancer cells to evade killing mediated by cytotoxic T lymphocytes. However, there are no studies on the immunological aspects of HDAC1 in non-small cell lung cancer (NSCLC). Methods In this research, we used the Cancer Genome Atlas (TCGA) public database combined with tissue microarray (TMA) to investigate HDAC1 expression and prognosis in NSCLC. According to the median value of HDAC1 expression in the TCGA dataset, samples of patients with NSCLC were classified into high- and low-expression cohorts. Subsequently, the biological characteristics of HDAC1 in high- and low-expression cohorts in terms of signaling pathways, immune cell infiltration, immune cell function, and genomic stability were investigated to better understand the effect of HDAC1 in the tumor microenvironment (TME) of NSCLC. Additionally, we selected tissue samples with HDAC1 overexpression in TMA2 and performed immunohistochemical staining of CD8+ T cells to observe the distribution of CD8+ T cells in the tumor. Results The findings revealed that overexpression of HDAC1 in NSCLC was associated with poor prognosis. Analysis of signaling pathway enrichment indicated that HDAC1 downregulated immune-related signaling pathways in NSCLC. Immune cell infiltration, immune cell function, and genomic stability analyses suggested that the TME alteration mediated by HDAC1 in the high-expression cohort was consistent with the "immune desert" phenotype. Furthermore, CD8+ T immunohistochemical staining experiments of tissue samples with HDAC1 overexpression in NSCLC revealed few CD8+ T cells distributed in the tumor parenchyma and interstitium. Conclusion Conclusively, our findings from several biological analyses revealed that HDAC1 is overexpressed in NSCLC and induces TME immunosuppression.
Collapse
Affiliation(s)
- Yongfei Fan
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Xiang Ji
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, People’s Republic of China
| | - Kai Yuan
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Qiyong Wu
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Ming Lou
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| |
Collapse
|
33
|
Ferry A, Mempel KM, Monell A, Reina-Campos M, Scharping NE, Heeg M, Takehara KK, Schokrpur S, Kuo N, Saddawi-Konefka R, Gutkind JS, Goldrath AW. The XCL1-XCR1 axis supports intestinal tissue residency and antitumor immunity. J Exp Med 2025; 222:e20240776. [PMID: 39841133 PMCID: PMC11753173 DOI: 10.1084/jem.20240776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/21/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Tissue-resident memory T cells (TRM) provide frontline protection against pathogens and emerging malignancies. Tumor-infiltrating lymphocytes (TIL) with TRM features are associated with improved clinical outcomes. However, the cellular interactions that program TRM differentiation and function are not well understood. Using murine genetic models and targeted spatial transcriptomics, we found that the CD8+ T cell-derived chemokine XCL1 is critical for TRM formation and conventional DC1 (cDC1) supported the positioning of intestinal CD8+ T cells during acute viral infection. In tumors, enforced Xcl1 expression by antigen-specific CD8+ T cells promoted intratumoral cDC1 accumulation and T cell persistence, leading to improved overall survival. Notably, analysis of human TIL and TRM revealed conserved expression of XCL1 and XCL2. Thus, we have shown that the XCL1-XCR1 axis plays a non-cell autonomous role in guiding intestinal CD8+ TRM spatial differentiation and tumor control.
Collapse
Affiliation(s)
- Amir Ferry
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kianoosh M. Mempel
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Alexander Monell
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Miguel Reina-Campos
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Nicole E. Scharping
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Maximilian Heeg
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kennidy K. Takehara
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Shiruyeh Schokrpur
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Ning Kuo
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | | | - J. Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Ananda W. Goldrath
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Allen Institute for Immunology, Seattle, WA, USA
| |
Collapse
|
34
|
Lukeman H, Al-Wassiti H, Fabb SA, Lim L, Wang T, Britton WJ, Steain M, Pouton CW, Triccas JA, Counoupas C. An LNP-mRNA vaccine modulates innate cell trafficking and promotes polyfunctional Th1 CD4 + T cell responses to enhance BCG-induced protective immunity against Mycobacterium tuberculosis. EBioMedicine 2025; 113:105599. [PMID: 39955975 PMCID: PMC11871481 DOI: 10.1016/j.ebiom.2025.105599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Mycobacterium tuberculosis remains the largest infectious cause of mortality worldwide, even with over a century of widespread administration of the only licenced tuberculosis (TB) vaccine, Bacillus Calmette-Guérin (BCG). mRNA technology remains an underexplored approach for combating chronic bacterial infections such as TB. METHODS We have developed a lipid nanoparticle (LNP)-mRNA vaccine, termed mRNACV2, encoding for the M. tuberculosis CysVac2 fusion protein, which we have previously formulated as an adjuvanted subunit vaccine. This LNP-mRNA vaccine was administered intramuscularly to female C57BL/6 mice as a standalone vaccine or as booster to BCG to assess immunogenicity and efficacy of the construct. FINDINGS Vaccination with mRNACV2 induced high frequencies of polyfunctional, antigen-specific Th1 CD4+ T cells in the blood and lungs, which was associated with the rapid recruitment of both innate and adaptive immune cells to lymph nodes draining the site of immunisation. mRNACV2 vaccination also provided significant pulmonary protection in M. tuberculosis-infected mice, reducing bacterial load and inflammatory infiltration in the lungs. Importantly, mRNACV2 enhanced immune responses and long-term protection when used to boost BCG-primed mice. INTERPRETATION These findings of a protective LNP-mRNA vaccine for TB highlight the potential of the LNP-mRNA platform for TB control and support further research to facilitate translation to humans. FUNDING This work was supported by the NHMRC Centre of Research Excellence in Tuberculosis Control to JAT and WJB (APP1153493), and MRFF mRNA Clinical Trial Enabling Infrastructure grant to CWP and HAW (MRFCTI000006).
Collapse
Affiliation(s)
- Hannah Lukeman
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Hareth Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Trixie Wang
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Warwick J Britton
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Megan Steain
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - James A Triccas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Claudio Counoupas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
35
|
Chua YC, Draper SL, Le S, de Menezes MN, Ganley M, Ge Z, Lee A, Phabmixay T, Hirschmann D, Robinson SA, Tan PS, Tullett KM, Anderson RJ, Jayasinghe D, Cozijnsen A, Lahoud MH, Caminschi I, Beattie L, McFadden GI, Larsen DS, Kaisho T, Gras S, Hermans IF, Compton BJ, Heath WR, Painter GF, Holz LE. Mechanistic insight into the induction of liver tissue-resident memory CD8 + T cells by glycolipid-peptide vaccination. Cell Rep 2025; 44:115295. [PMID: 39946236 DOI: 10.1016/j.celrep.2025.115295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 10/12/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
We recently demonstrated that vaccines comprising antigenic peptides conjugated to a glycolipid agonist, termed glycolipid-peptide (GLP) vaccines, efficiently generate substantial numbers of long-lived CD8+ liver-resident memory T (Trm) cells that are crucial for protection against malaria liver-stage infection. To understand the underlying mechanism, we examined the prerequisites for priming, differentiation, and secondary boosting of liver Trm cells using these GLP vaccines. Our study revealed that generation of long-lived liver Trm cells relies on CD8+ T cell priming by type 1 conventional dendritic (cDC1) cells, followed by post-priming exposure to a combination of vaccine-derived inflammatory and antigenic signals. Boosting of liver Trm cells is feasible using the same GLP vaccine, but a substantial delay is required for optimal responses due to natural killer T (NKT) cell anergy. Overall, our study unveils key requirements for the development of long-lived liver Trm cells, offering valuable insights for future vaccine design.
Collapse
Affiliation(s)
- Yu Cheng Chua
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Sarah L Draper
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand
| | - Shirley Le
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Maria N de Menezes
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Mitch Ganley
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Zhengyu Ge
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Ariane Lee
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Taylah Phabmixay
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Daria Hirschmann
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Sage A Robinson
- Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Peck Szee Tan
- Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3168, Australia
| | - Kirsteen M Tullett
- Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3168, Australia
| | - Regan J Anderson
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand
| | - Dhilshan Jayasinghe
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
| | - Anton Cozijnsen
- School of BioSciences, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Mireille H Lahoud
- Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3168, Australia
| | - Irina Caminschi
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3168, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Geoffrey I McFadden
- School of BioSciences, The University of Melbourne, Parkville, VIC 3052, Australia
| | - David S Larsen
- Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Stephanie Gras
- Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3168, Australia; Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand
| | - William R Heath
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand
| | - Lauren E Holz
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia.
| |
Collapse
|
36
|
Huang L, Liu Y, Shi Y, Sun Q, Li H, Sun C. Comprehensive single-cell analysis of triple-negative breast cancer based on cDC1 immune-related genes: prognostic model construction and immunotherapy potential. Discov Oncol 2025; 16:206. [PMID: 39969635 PMCID: PMC11839968 DOI: 10.1007/s12672-025-01929-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Various components of the immunological milieu surrounding tumors have become a key focus in cancer immunotherapy research. There are currently no reliable biomarkers for triple-negative breast cancer (TNBC), leading to limited clinical benefits. However, some studies have indicated that patients with TNBC may achieve better outcomes after immunotherapy. Therefore, this study aimed to identify molecular features potentially associated with conventional type 1 dendritic cell (cDC1) immunity to provide new insights into TNBC prognostication and immunotherapy decision-making. METHODS Single-cell ribonucleic acid sequencing data from the Gene Expression Omnibus database were analyzed to determine which genes are differentially expressed genes (DEGs) in cDC1s. We then cross-referenced cDC1-related DEGs with gene sets linked to immunity from the ImmPort and InnateDB databases to screen for the genes linked to the immune response and cDC1s. We used univariate Cox and least absolute shrinkage and selection operator regression analyses to construct a risk assessment model based on four genes in patients with TNBC obtained from the Cancer Genome Atlas, which was validated in a testing group. This model was also used to assess immunotherapy responses among the IMvigor210 cohort. We subsequently utilized single sample Gene Set Enrichment Analysis, CIBERSORT, and ESTIMATE to analyze the immunological characteristics of the feature genes and their correlation with drug response. RESULTS We identified 93 DEGs related to the immune response and cDC1s, of which four (IDO1, HLA-DOB, CTSD, and IL3RA) were substantially linked to the overall survival rate of TNBC patients. The risk assessment model based on these genes stratified patients into high- and low-risk groups. Low-risk patients exhibited enriched ''hot tumor'' phenotypes, including higher infiltration of memory-activated CD4 + T cells, CD8 + T cells, gamma delta T cells, and M1 macrophages, as well as elevated immune checkpoint expression and tumor mutational burden, suggesting potential responsiveness to immunotherapy. Conversely, high-risk patients displayed "cold tumor" characteristics, with higher infiltration of M0 and M2 macrophages and lower immune scores, which may be poorer in response to immunotherapy. However, experimental validation and larger clinical studies are necessary to confirm these findings and explore the underlying mechanisms of the identified genes. CONCLUSION This study developed a robust risk assessment model using four genes that effectively forecast the outcome of patients with TNBC and have the potential to guide immunotherapy. This model provided new theoretical insights for knowing the TNBC immune microenvironment and developing personalized treatment strategies.
Collapse
Affiliation(s)
- Linan Huang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China
| | - Yiran Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China
| | - Yulin Shi
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| |
Collapse
|
37
|
Mao H, Dumas EK, Starnbach MN. Chlamydia trachomatis impairs T cell priming by inducing dendritic cell death. Infect Immun 2025; 93:e0040224. [PMID: 39772728 PMCID: PMC11834465 DOI: 10.1128/iai.00402-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
The lack of effective adaptive immunity against Chlamydia trachomatis leads to chronic or repeated infection and serious disease sequelae. Dendritic cells (DCs) are professional antigen-presenting cells that are crucial for the activation of T cells during C. trachomatis infection. cDC1s and cDC2s are the two main DC subsets responsible for T cell priming, but little is known about how C. trachomatis affects their ability to prime T cells. Using a mouse model of infection, we found that C. trachomatis uptake reduced the viability of cDC1s and cDC2s both in vitro and in vivo, with cDC1s experiencing more death. DC death was mainly due to apoptosis and is alleviated in Casp3/7 or Bak1/Bax knockout DCs. In addition, we observed that C. trachomatis-specific CD8+ T cells were preferentially activated by cDC1s. Reduction in DC viability by C. trachomatis impaired the ability of infected DCs to activate T cells upon co-culture, although in the case of CD8+ T cell priming, controlling for viability was insufficient to fully rescue the defect. RNA sequencing of DCs from infected mice showed upregulation of cell death pathways, supporting our observations of DC death caused by C. trachomatis. Finally, we validated our findings with human DCs in vitro, observing C. trachomatis-induced cell death. These results indicate that C. trachomatis may evade the adaptive immune system by directly inducing cell death in DCs.
Collapse
Affiliation(s)
- Haitong Mao
- />Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric K. Dumas
- />Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael N. Starnbach
- />Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Carter H, Costa RM, Adams TS, Gilchrist TM, Emch CE, Bame M, Oldham JM, Huang SK, Linderholm AL, Noth I, Kaminski N, Moore BB, Gurczynski SJ. CD103+ dendritic cell-fibroblast crosstalk via TLR9, TDO2, and AHR signaling drives lung fibrogenesis. JCI Insight 2025; 10:e177072. [PMID: 39964756 PMCID: PMC11949071 DOI: 10.1172/jci.insight.177072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive scarring and loss of lung function. With limited treatment options, patients die from the disease within 2-5 years. The molecular pathogenesis underlying the immunologic changes that occur in IPF is poorly understood. We characterize noncanonical aryl-hydrocarbon receptor (ncAHR) signaling in DCs as playing a role in the production of IL-6 and increased IL-17+ cells, promoting fibrosis. TLR9 signaling in myofibroblasts is shown to regulate production of TDO2, which converts tryptophan into the endogenous AHR ligand kynurenine. Mice with augmented ncAHR signaling were created by crossing mice harboring a floxed AHR exon 2 deletion (AHRΔex2) with mice harboring a CD11c-Cre. Bleomycin (blm) was used to study fibrotic pathogenesis. Isolated CD11c+ cells and primary fibroblasts were treated ex vivo with relevant TLR agonists and AHR-modulating compounds to study how AHR signaling influenced inflammatory cytokine production. Human datasets were also interrogated. Inhibition of all AHR signaling rescued fibrosis; however, AHRΔex2 mice treated with blm developed more fibrosis, and DCs from these mice were hyperinflammatory and profibrotic upon adoptive transfer. Treatment of fibrotic fibroblasts with TLR9 agonist increased expression of TDO2, and fibrotic fibroblasts activated IL-6 production in CD103+ DCs. Study of human samples corroborated the relevance of these findings in patients with IPF. We also show, for the first time to our knowledge, that AHR exon 2 floxed mice retain the capacity for ncAHR signaling.
Collapse
Affiliation(s)
- Hannah Carter
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rita Medina Costa
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Talon M. Gilchrist
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Claire E. Emch
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Steven K. Huang
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Angela L. Linderholm
- Division of Pulmonary and Critical Care Medicine, University of California, Davis, California, USA
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephen J. Gurczynski
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
39
|
Qin D, Lei Y, Shu P, Zhang Y, Loh YH, Wang Y, Li Q. Supercharging CAR-T cells through transcriptional and epigenetic armoring. Theranostics 2025; 15:3345-3367. [PMID: 40093905 PMCID: PMC11905144 DOI: 10.7150/thno.107908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Inspired by the remarkable success of CAR-T therapy in hematologic malignancies, research is increasingly focused on adapting this treatment for solid tumors. However, CAR-T efficacy remains limited due to its exhaustion and shortened persistence. Transcription factors and epigenetic modifications play pivotal roles in modulating T cell differentiation and functionality, which have been leveraged in numerous strategies to promote the formation of long-lasting memory cells with stem-like properties and supercharging CAR-T performance. This review highlights pivotal transcriptional factors, such as c-Jun and FOXO1, which enhance and sustain T cell effector function, diminishes exhaustion, and epigenetic regulators like TET2 and DNMT3A, whose knockout promotes memory T subsets formation. We explore their interconnections, downstream targets, biological impacts, and the potential application risks of certain candidates, providing a comprehensive theoretical framework for supercharging CAR-T therapies through transcriptional and epigenetic interventions.
Collapse
Affiliation(s)
- Diyuan Qin
- Cancer Center, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
| | - Yanna Lei
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pei Shu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yugu Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Cancer Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qijing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A ∗ STAR), Singapore 138673, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
| |
Collapse
|
40
|
Hussain Z, Zhang Y, Qiu L, Gou S, Liu K. Exploring Clec9a in dendritic cell-based tumor immunotherapy for molecular insights and therapeutic potentials. NPJ Vaccines 2025; 10:27. [PMID: 39920156 PMCID: PMC11806010 DOI: 10.1038/s41541-025-01084-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
The pivotal role of type 1 conventional dendritic cells (cDC1s) in the field of dendritic cell (DC)-based tumor immunotherapies has been gaining increasing recognition due to their superior antigen cross-presentation abilities and essential role in modulating immune responses. This review specifically highlights the C-type lectin receptor family 9 member A (Clec9a or DNGR-1), which is exclusively expressed on cDC1s and plays a pivotal role in augmenting antigen cross-presentation and cytotoxic T lymphocyte (CTL) responses while simultaneously mitigating off-target effects. These effects include the enhancement of the cDC1s cross-presentation, reducing autoimmune responses and systemic inflammation, as well as preventing the non-specific activation of other immune cells. Consequently, these actions may contribute to reduced toxicity and enhanced treatment efficacy in immunotherapy. The exceptional ability of Clec9a to cross-present dead cell-associated antigens and enhance both humoral and CTL responses makes it an optimal receptor for DC-based strategies aimed at strengthening antitumor immunity. This review provides a comprehensive overview of the molecular characterization, expression, and signaling mechanisms of Clec9a. Furthermore, it discusses the role of Clec9a in the induction and functional activation of Clec9a+ cDC1s, with a particular focus on addressing the challenges related to off-target effects and immune tolerance in the development of tumor vaccines. Additionally, this review explores the potential of Clec9a-targeted approaches to enhance the immunogenicity of tumor vaccines and addresses the utilization of Clec9a as a delivery target for specific agonists (such as STING agonists and αGC) to enhance their therapeutic effects. This novel approach leverages Clec9a's capacity to improve the precision and efficacy of these immunomodulatory molecules in tumor treatment. In summary, this review presents compelling evidence positioning Clec9a as a promising target for DC-based tumor immunotherapy, capable of enhancing the efficacy of vaccines and immune responses while minimizing adverse effects.
Collapse
Affiliation(s)
- Zubair Hussain
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yueteng Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Qiu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Gou
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
- China‒US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China.
| |
Collapse
|
41
|
Morita S, Lei PJ, Shigeta K, Ando T, Kobayashi T, Kikuchi H, Matsui A, Huang P, Pittet MJ, Duda DG. Combination CXCR4 and PD-1 Blockade Enhances Intratumoral Dendritic Cell Activation and Immune Responses Against Hepatocellular Carcinoma. Cancer Immunol Res 2025; 13:162-170. [PMID: 39514263 PMCID: PMC11788650 DOI: 10.1158/2326-6066.cir-24-0324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Immune checkpoint inhibitors have revolutionized the treatment of unresectable hepatocellular carcinoma (HCC), but their impressive efficacy is seen in just a fraction of patients. One key mechanism of immunotherapy resistance is the paucity of dendritic cells (DC) in liver malignancies. In this study, we tested combination blockade of PD-1 and CXCR4, a receptor for CXCL12, a pleiotropic factor that mediates immunosuppression in tumors. Using orthotopic grafted and autochthonous HCC models with underlying liver damage, we evaluated treatment feasibility and efficacy. In addition, we examined the effects of treatment using immunofluorescence, flow cytometric analysis of DCs in vivo and in vitro, and RNA sequencing. The combination anti-CXCR4 and anti-PD-1 therapy was safe and significantly inhibited tumor growth and prolonged survival in all murine preclinical models of HCC tested. The combination treatment successfully reprogrammed antigen-presenting cells, revealing the potential role of conventional type 1 DCs (cDC1) in the HCC microenvironment. Moreover, DC reprogramming enhanced anticancer immunity by facilitating CD8+ T-cell accumulation and activation in the HCC tissue. The effectiveness of anti-CXCR4/PD-1 therapy was compromised entirely in Batf3 knockout mice deficient in cDC1s. Thus, combined CXCR4/PD-1 blockade can reprogram intratumoral cDC1s and holds the potential to potentiate antitumor immune response against HCC.
Collapse
Affiliation(s)
- Satoru Morita
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Pin-Ji Lei
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kohei Shigeta
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tomofumi Ando
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuya Kobayashi
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hiroto Kikuchi
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Aya Matsui
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Kanazawa University Institute of Medical, Pharmaceutical and Health Sciences Faculty of Medicine, Kanazawa, Japan
| | - Peigen Huang
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mikael J. Pittet
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Dan G. Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Tuor M, Stappers MHT, Desgardin A, Ruchti F, Sparber F, Orr SJ, Gow NAR, LeibundGut-Landmann S. Card9 and MyD88 differentially regulate Th17 immunity to the commensal yeast Malassezia in the murine skin. Mucosal Immunol 2025; 18:205-219. [PMID: 39579986 DOI: 10.1016/j.mucimm.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
The fungal community of the skin microbiome is dominated by a single genus, Malassezia. Besides its symbiotic lifestyle at the host interface, this commensal yeast has also been associated with diverse inflammatory skin diseases in humans and pet animals. Stable colonization is maintained by antifungal type 17 immunity. The mechanisms driving Th17 responses to Malassezia remain, however, unclear. Here, we show that the C-type lectin receptors Mincle, Dectin-1, and Dectin-2 recognize conserved patterns in the cell wall of Malassezia and induce dendritic cell activation in vitro, while only Dectin-2 is required for Th17 activation during experimental skin colonization in vivo. In contrast, Toll-like receptor recognition was redundant in this context. Instead, inflammatory IL-1 family cytokines signaling via MyD88 were also implicated in Th17 activation in a T cell-intrinsic manner. Taken together, we characterized the pathways contributing to protective immunity against the most abundant member of the skin mycobiome. This knowledge contributes to the understanding of barrier immunity and its regulation by commensals and is relevant considering how aberrant immune responses are associated with severe skin pathologies.
Collapse
Affiliation(s)
- Meret Tuor
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Mark H T Stappers
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Alice Desgardin
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Florian Sparber
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Selinda J Orr
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland; Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK.
| |
Collapse
|
43
|
Zhou J, Xiong KL, Wang HX, Sun WW, Ke H, Zhang SJ, Dong ZW, Fan L. BATF2/SINHCAF regulates the quantity and function of macrophages infected with Mycobacterium Tuberculosis via regulation of TTC23 through Wnt/β-catenin pathway. Int J Biol Macromol 2025; 288:138639. [PMID: 39672395 DOI: 10.1016/j.ijbiomac.2024.138639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Elucidating the pathogenic mechanism of Tuberculosis (TB) can contribute to control TB. Basic leucine zipper transcription factor ATF-like 2 (BATF2) belonging to a large family of leucine zipper transcription factors (TFs) termed bZip proteins, had been verified to have important value in the diagnosis of TB. However, its role and mechanism in TB had not been elucidated. The study aimed to explore its function and molecular mechanism in macrophages infected with Mycobacterium tuberculosis (Mtb). The results indicated that BATF2 inhibited cell proliferation, promoted inflammatory response and impaired the antibacterial and antigen-presenting capacity in macrophages for T cells through regulating its downstream gene TTC23 by interacting with SINHCAF. Above roles and regulations were dependent on β-catenin functions in macrophages infected with Mtb. Clinical samples verified that the expressions of BATF2 and TTC23 were significantly higher in the blood of patients with pulmonary TB compared with health controls. Altogether, BATF2 interacted with SINHCAF to regulate the quantity and function of macrophages during Mtb infection by targeting TTC23 through Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Kun-Long Xiong
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Hong-Xiu Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Wen Sun
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Hui Ke
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Shao-Jun Zhang
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Zheng-Wei Dong
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Fan
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China.
| |
Collapse
|
44
|
Sebastião AI, Simões G, Oliveira F, Mateus D, Falcão A, Carrascal MA, Gomes C, Neves B, Cruz MT. Dendritic cells in triple-negative breast cancer: From pathophysiology to therapeutic applications. Cancer Treat Rev 2025; 133:102884. [PMID: 39837068 DOI: 10.1016/j.ctrv.2025.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/28/2024] [Accepted: 01/11/2025] [Indexed: 01/23/2025]
Abstract
Breast cancer is the second most commonly diagnosed cancer in women and the fifth leading cause of cancer-related deaths worldwide. It is a highly heterogeneous disease, consisting of multiple subtypes that vary significantly in clinical characteristics and survival outcomes. Triple-negative breast cancer (TNBC) is a particularly aggressive and challenging subtype of breast cancer. Several immunotherapeutic approaches have been tested in patients with TNBC to improve disease outcomes, including the administration of dendritic cell (DC)-based vaccines. DCs are a heterogeneous cell population that play a crucial role in bridging the innate and adaptive immune systems. Therefore, DCs have been increasingly used in cancer vaccines due to their ability to prime and boost antigen specific T-cell immune responses. This review aims to provide a comprehensive overview of TNBC, including potential targets and pharmacological strategies, as well as an overview of DCs and their relevance in TNBC. In addition, we review ongoing clinical trials and shed light on the evolving landscape of DC-based immunotherapy for TNBC.
Collapse
Affiliation(s)
- Ana Isabel Sebastião
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Gonçalo Simões
- Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Filomena Oliveira
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Mateus
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; BioMark@UC/CEB-LABBELS, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
| | | | - Célia Gomes
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research - iCBR, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Bruno Neves
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
45
|
León B. Type 2 conventional dendritic cell functional heterogeneity: ontogenically committed or environmentally plastic? Trends Immunol 2025; 46:104-120. [PMID: 39843310 PMCID: PMC11835539 DOI: 10.1016/j.it.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/24/2025]
Abstract
Conventional dendritic cells (cDCs) are sentinels of the mammalian immune system that sense a wide range of danger and homeostatic signals to induce appropriately targeted T cell immune responses. Traditionally classified into two main subsets, cDC1 and cDC2, recent research shows that cDC2s exhibit significant heterogeneity and can be further subdivided. Studies in mice and humans show that, beyond their ontogeny, cDC2s acquire dynamic and tissue-specific characteristics that are influenced by local environmental signals, which impact on their functions during homeostasis, inflammation, and infection. The novel concept is proposed that tissue-derived signals and tissue plasticity can override preestablished developmental programming, thereby redefining developmental trajectories and cDC2 functionality. Ultimately, understanding cDC2 heterogeneity and plasticity has important implications for modulating T cell immunity in health and disease.
Collapse
Affiliation(s)
- Beatriz León
- Innate Cells and Th2 Immunity Section, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
46
|
Chang H, Li M, Zhang L, Li M, Ong SH, Zhang Z, Zheng J, Xu X, Zhang Y, Wang J, Liu X, Li K, Luo Y, Wang H, Miao Z, Chen X, Zha J, Yu Y. Loss of histone deubiquitinase Bap1 triggers anti-tumor immunity. Cell Oncol (Dordr) 2025; 48:183-203. [PMID: 39141316 PMCID: PMC11850471 DOI: 10.1007/s13402-024-00978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Immunotherapy using PD-L1 blockade is effective in only a small group of cancer patients, and resistance is common. This emphasizes the importance of understanding the mechanisms of cancer immune evasion and resistance. METHODS A genome-scale CRISPR-Cas9 screen identified Bap1 as a regulator of PD-L1 expression. To measure tumor size and survival, tumor cells were subcutaneously injected into both syngeneic WT mice and immunocompromised mice. The phenotypic and transcriptional characteristics of Bap1-deleted tumors were examined using flow cytometry, RNA-seq, and CUT&Tag-seq analysis. RESULTS We found that loss of histone deubiquitinase Bap1 in cancer cells activates a cDC1-CD8+ T cell-dependent anti-tumor immunity. The absence of Bap1 leads to an increase in genes associated with anti-tumor immune response and a decrease in genes related to immune evasion. As a result, the tumor microenvironment becomes inflamed, with more cDC1 cells and effector CD8+ T cells, but fewer neutrophils and regulatory T cells. We also found that the elimination of Bap1-deleted tumors depends on the tumor MHCI molecule and Fas-mediated CD8+ T cell cytotoxicity. Our analysis of TCGA data further supports these findings, showing a reverse correlation between BAP1 expression and mRNA signatures of activated DCs and T-cell cytotoxicity in various human cancers. CONCLUSION The histone deubiquitinase Bap1 could be used as a biomarker for tumor stratification and as a potential therapeutic target for cancer immunotherapies.
Collapse
Affiliation(s)
- Hong Chang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Mingxia Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Linlin Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Meng Li
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Swee Hoe Ong
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Zhiwei Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jie Zheng
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xiang Xu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jing Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xingjie Liu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Kairui Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yao Luo
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Haiyun Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhichao Miao
- Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Xi Chen
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, China.
| | - Yong Yu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
47
|
Wang X, Hong Y, Zou J, Zhu B, Jiang C, Lu L, Tian J, Yang J, Rui K. The role of BATF in immune cell differentiation and autoimmune diseases. Biomark Res 2025; 13:22. [PMID: 39876010 PMCID: PMC11776340 DOI: 10.1186/s40364-025-00733-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
As a member of the Activator Protein-1 (AP-1) transcription factor family, the Basic Leucine Zipper Transcription Factor (BATF) mediates multiple biological functions of immune cells through its involvement in protein interactions and binding to DNA. Recent studies have demonstrated that BATF not only plays pivotal roles in innate and adaptive immune responses but also acts as a crucial factor in the differentiation and function of various immune cells. Lines of evidence indicate that BATF is associated with the onset and progression of allergic diseases, graft-versus-host disease, tumors, and autoimmune diseases. This review summarizes the roles of BATF in the development and function of innate and adaptive immune cells, as well as its immunoregulatory effects in the development of autoimmune diseases, which may enhance the current understanding of the pathogenesis of autoimmune diseases and facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Department of Laboratory Medicine, Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yue Hong
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jinmei Zou
- Department of Rheumatology, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China
| | - Bo Zhu
- Department of Laboratory Medicine, Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chao Jiang
- Department of Orthopaedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Liwei Lu
- Department of Pathology, Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Jing Yang
- Department of Rheumatology, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, China.
| | - Ke Rui
- Department of Laboratory Medicine, Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
48
|
Bandola-Simon J, Ito Y, Wucherpfennig KW, Roche PA. Defective removal of invariant chain peptides from MHC class II suppresses tumor antigen presentation and promotes tumor growth. Cell Rep 2025; 44:115150. [PMID: 39752250 PMCID: PMC11886875 DOI: 10.1016/j.celrep.2024.115150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 02/01/2025] Open
Abstract
Tumor-draining lymph node dendritic cells (DCs) are poor stimulators of tumor antigen-specific CD4 T cells; however, the mechanism behind this defect is unclear. We now show that, in tumor-draining lymph node DCs, a large proportion of major histocompatibility complex class II (MHC-II) molecules retains the class II-associated invariant chain peptide (CLIP) fragment of the invariant chain bound to the MHC-II peptide binding groove due to reduced expression of the peptide editor H2-M and enhanced activity of the CLIP-generating proteinase cathepsin S. The net effect of this is that MHC-II molecules are unable to efficiently bind antigenic peptides. DCs in mice expressing a mutation in the invariant chain sequence that results in enhanced MHC-II-CLIP accumulation are poor stimulators of CD4 T cells and have diminished anti-tumor responses. Our data reveal a previously unknown mechanism of immune evasion in which enhanced expression of MHC-II-CLIP complexes on tumor-draining lymph node DCs limits MHC-II availability for tumor peptides.
Collapse
MESH Headings
- Histocompatibility Antigens Class II/metabolism
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/genetics
- Animals
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigen Presentation/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Mice
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Mice, Inbred C57BL
- CD4-Positive T-Lymphocytes/immunology
- Peptides/metabolism
- Peptides/immunology
- Lymph Nodes/immunology
- Neoplasms/immunology
- Neoplasms/pathology
- Cell Line, Tumor
- Humans
Collapse
Affiliation(s)
- Joanna Bandola-Simon
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshinaga Ito
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
49
|
Huis In 't Veld LG, Cornelissen LA, van den Bogaard L, Ansems M, Ho NI, Adema GJ. Saponin-based adjuvant uptake and induction of antigen cross-presentation by CD11b+ dendritic cells and macrophages. NPJ Vaccines 2025; 10:15. [PMID: 39843492 PMCID: PMC11754886 DOI: 10.1038/s41541-024-01056-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Saponin-based adjuvants (SBAs) distinguish themselves as vaccine adjuvants by instigating a potent activation of CD8+ T cells. Previously, we discovered SBA's ability to induce cross-presentation in dendritic cells (DCs) leading to CD8+ T cell activation. Moreover, the MHCIIloCD11bhi bone marrow-derived DC (BMDC) subset was identified to be the most responsive DC subset to SBA treatment. To further investigate SBA's mode of action, labeling of SBAs was optimized with the fluorescent dye SP-DiIC18(3). Efficient uptake of SBAs occurs specifically by MHCIIloCD11bhi BMDCs and bone marrow-derived macrophages (BMDMs) in vitro and cDC2s and macrophages ex vivo. Furthermore, SBAs are primarily taken up by clathrin-mediated endocytosis and uptake induces lipid bodies and antigen translocation to the cytosol in MHCIIloCD11bhi BMDCs and BMDMs. Importantly, BMDMs treated with SBAs exhibit cross-presentation leading to potent CD8+ T cells activation. Our findings explain the potency of SBAs as vaccine adjuvants and contribute to vaccine development.
Collapse
Affiliation(s)
- Lisa Gm Huis In 't Veld
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lenneke Am Cornelissen
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lune van den Bogaard
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nataschja I Ho
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
50
|
Cook PC, Brown SL, Houlder EL, Furlong-Silva J, Conn DP, Colombo SAP, Baker S, Svedberg FR, Howell G, Bertuzzi M, Boon L, Konkel JE, Thornton CR, Allen JE, MacDonald AS. Mgl2 + cDC2s coordinate fungal allergic airway type 2, but not type 17, inflammation in mice. Nat Commun 2025; 16:928. [PMID: 39843887 PMCID: PMC11754877 DOI: 10.1038/s41467-024-55663-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025] Open
Abstract
Fungal spores are abundant in the environment and a major cause of asthma. Originally characterised as a type 2 inflammatory disease, allergic airway inflammation that underpins asthma can also involve type 17 inflammation, which can exacerbate disease causing failure of treatments tailored to inhibit type 2 factors. However, the mechanisms that determine the host response to fungi, which can trigger both type 2 and type 17 inflammation in allergic airway disease, remain unclear. Here we find that CD11c+ DCs and CD4+ T cells are essential for development of both type 2 and type 17 airway inflammation in mice repeatedly exposed to inhaled spores. Single cell RNA-sequencing with further multi-parameter cytometry shows that allergic inflammation dramatically alters the proportion of numerous DC clusters in the lung, but that only two of these (Mgl2+ cDC2s and CCR7+ DCs) migrate to the dLNs. Targeted removal of several DC subsets shows that Mgl2+ cDC2 depletion reduces type 2, but not type 17, fungal allergic airway inflammation. These data highlight distinct DC subsets as potential therapeutic targets for the treatment of pulmonary fungal disease.
Collapse
Affiliation(s)
- Peter C Cook
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter, United Kingdom.
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.
| | - Sheila L Brown
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Emma L Houlder
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Julio Furlong-Silva
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter, United Kingdom
| | - Daniel P Conn
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter, United Kingdom
| | - Stefano A P Colombo
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Syed Baker
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Freya R Svedberg
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Gareth Howell
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Margherita Bertuzzi
- Manchester Fungal Infection Group, University of Manchester, Manchester, United Kingdom
| | | | - Joanne E Konkel
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christopher R Thornton
- Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Judith E Allen
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|