1
|
Aljobaily N, Allard D, Perkins B, Raugh A, Galland T, Jing Y, Stephens WZ, Bettini ML, Hale JS, Bettini M. Autoimmune CD4 + T cells fine-tune TCF1 expression to maintain function and survive persistent antigen exposure during diabetes. Immunity 2024; 57:2583-2596.e6. [PMID: 39396521 DOI: 10.1016/j.immuni.2024.09.016] [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: 05/09/2024] [Revised: 07/21/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
Self-reactive T cells experience chronic antigen exposure but do not exhibit signs of exhaustion. Here, we investigated the mechanisms for sustained, functioning autoimmune CD4+ T cells despite chronic stimulation. Examination of T cell priming showed that CD4+ T cells activated in the absence of infectious signals retained TCF1 expression. At later time points and during blockade of new T cell recruitment, most islet-infiltrating autoimmune CD4+ T cells were TCF1+, although expression was reduced on a per T cell basis. The Tcf7 locus was epigenetically modified in circulating autoimmune CD4+ T cells, suggesting a pre-programmed de novo methylation of the locus in early stages of autoimmune CD4+ T cell differentiation. This mirrored the epigenetic profile of recently recruited CD4+CD62L+ T cells in the pancreas. Collectively, these data reveal a unique environment during autoimmune CD4+ T cell priming that allows T cells to fine-tune TCF1 expression and maintain long-term survival and function.
Collapse
Affiliation(s)
- Nouf Aljobaily
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Denise Allard
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Bryant Perkins
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Arielle Raugh
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Tessa Galland
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Yi Jing
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - W Zac Stephens
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Matthew L Bettini
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - J Scott Hale
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Maria Bettini
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
2
|
Wang B, Bian Q. SATB1 prevents immune cell infiltration by regulating chromatin organization and gene expression of a chemokine gene cluster in T cells. Commun Biol 2024; 7:1304. [PMID: 39394451 PMCID: PMC11470149 DOI: 10.1038/s42003-024-07021-8] [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: 03/14/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024] Open
Abstract
SATB1, a key regulator of T cell development, governs lineage-specific transcriptional programs upon T cell activation. The absence of SATB1 has been linked to the initiation and progression of autoimmunity. However, its precise roles in this process remain incompletely understood. Here we show that conditional knockout of Satb1 in CD4+ T cells in mice led to T cell hyperactivation and inflammatory cell infiltration across multiple organs. Transcriptional profiling on activated T cells revealed that the loss of SATB1 led to aberrant upregulation of CC chemokines. Treating Satb1 conditional knockout mice with CC chemokine receptor inhibitor alleviated inflammatory cell infiltration. Intriguingly, SATB1's transcriptional regulation of chemokine genes could not be attributed to its direct binding to chemokine promoters. Instead, SATB1 exerted its regulatory effects by controlling higher-order chromatin organization at a CC chemokine locus. The loss of SATB1 led to the emergence of a new chromatin domain encompassing the Ccl3, Ccl4, Ccl5, Ccl6, and Ccl9 genes and a distal enhancer, resulting in increased contacts between the enhancer and all five chemokine genes, thus inducing their upregulation. Collectively, these results demonstrate that SATB1 protects organs from immune cell infiltration by regulating chemokine expression, providing valuable insights into the development of autoimmunity-related phenotypes.
Collapse
Affiliation(s)
- Bao Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Bromley JD, Ganchua SKC, Nyquist SK, Maiello P, Chao M, Borish HJ, Rodgers M, Tomko J, Kracinovsky K, Mugahid D, Nguyen S, Wang QD, Rosenberg JM, Klein EC, Gideon HP, Floyd-O'Sullivan R, Berger B, Scanga CA, Lin PL, Fortune SM, Shalek AK, Flynn JL. CD4 + T cells re-wire granuloma cellularity and regulatory networks to promote immunomodulation following Mtb reinfection. Immunity 2024; 57:2380-2398.e6. [PMID: 39214090 PMCID: PMC11466276 DOI: 10.1016/j.immuni.2024.08.002] [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: 12/22/2023] [Revised: 06/03/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Immunological priming-in the context of either prior infection or vaccination-elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrated that prior Mtb infection elicited a long-lasting protective response against subsequent Mtb exposure and was CD4+ T cell dependent. By analyzing data from primary infection, reinfection, and reinfection-CD4+ T cell-depleted granulomas, we found that the presence of CD4+ T cells during reinfection resulted in a less inflammatory lung milieu characterized by reprogrammed CD8+ T cells, reduced neutrophilia, and blunted type 1 immune signaling among myeloid cells. These results open avenues for developing vaccines and therapeutics that not only target lymphocytes but also modulate innate immune cells to limit tuberculosis (TB) disease.
Collapse
Affiliation(s)
- Joshua D Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharie Keanne C Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah K Nyquist
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Chao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - H Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Douaa Mugahid
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Son Nguyen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Qianchang Dennis Wang
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob M Rosenberg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edwin C Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah P Gideon
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roisin Floyd-O'Sullivan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah M Fortune
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Sirivisoot S, Kasantikul T, Techangamsuwan S, Rungsipipat A. Single nucleotide polymorphism profiles of canine T-cell and null-cell lymphomas. Front Vet Sci 2024; 11:1439706. [PMID: 39176397 PMCID: PMC11339873 DOI: 10.3389/fvets.2024.1439706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Background The histopathological classification of T-cell lymphoma (TCL) in humans has distinctive mutational genotyping that suggests different lymphomagenesis. A similar concept is assumed to be observed in dogs with different TCL phenotypes. Objective This study aimed to identify the previously reported single-nucleotide polymorphisms (SNPs) in both human beings and dogs in canine TCLs and null-cell lymphomas (NCLs) and to design compatible oligonucleotides from each variant based on the multiplex polymerase chain reaction. Methods Genomic DNA was extracted from 68 tumor specimens (62 TCLs and 6 NCLs) and 5 buffy coat samples from dogs with TCL. Four TCL subtypes and NCL were analyzed in 44 SNPs from 21 genes using the MassARRAY. Results The greatest incidences of SNPs observed in all TCL subtypes and NCL ware SATB1 c.1259A > C, KIT c.1275A > G, SEL1L c.2040 + 200C > G, and TP53 c.1024C > T, respectively. Some SNP locations were statistically significant associated with NCL, including MYC p.S75F (p = 0.0003), TP53 p.I149N (p = 0.030), PDCD1 p.F37LX (p = 0.012), and POT1 p.R583* (p = 0.012). Conclusion Each TCL histological subtype and NCL are likely to contain distinctive mutational genetic profiles, which might play a role in lymphoma gene-risk factors and might be useful for selecting therapeutic target drugs for each canine patient.
Collapse
Affiliation(s)
- Sirintra Sirivisoot
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Tanit Kasantikul
- Veterinary Diagnostic Laboratory, Michigan State University, Lansing, MI, United States
| | - Somporn Techangamsuwan
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Anudep Rungsipipat
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Qi W, Bai J, Wang R, Zeng X, Zhang L. SATB1, senescence and senescence-related diseases. J Cell Physiol 2024; 239:e31327. [PMID: 38801120 DOI: 10.1002/jcp.31327] [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: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Aging leads to an accumulation of cellular mutations and damage, increasing the risk of senescence, apoptosis, and malignant transformation. Cellular senescence, which is pivotal in aging, acts as both a guard against cellular transformation and as a check against cancer progression. It is marked by stable cell cycle arrest, widespread macromolecular changes, a pro-inflammatory profile, and altered gene expression. However, it remains to be determined whether these differing subsets of senescent cells result from unique intrinsic programs or are influenced by their environmental contexts. Multiple transcription regulators and chromatin modifiers contribute to these alterations. Special AT-rich sequence-binding protein 1 (SATB1) stands out as a crucial regulator in this process, orchestrating gene expression by structuring chromatin into loop domains and anchoring DNA elements. This review provides an overview of cellular senescence and delves into the role of SATB1 in senescence-related diseases. It highlights SATB1's potential in developing antiaging and anticancer strategies, potentially contributing to improved quality of life and addressing aging-related diseases.
Collapse
Affiliation(s)
- Wenjing Qi
- Department of Bioscience, Changchun Normal University, Changchun, Jilin, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, Jilin, China
| | - Jinping Bai
- Department of Bioscience, Changchun Normal University, Changchun, Jilin, China
| | - Ruoxi Wang
- Center for Cell Structure and Function, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, Shandong, China
| | - Xianlu Zeng
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, Jilin, China
| | - Lihui Zhang
- Department of Bioscience, Changchun Normal University, Changchun, Jilin, China
| |
Collapse
|
6
|
Liang L, Yang Y, Deng K, Wu Y, Li Y, Bai L, Wang Y, Lu C. Type I Interferon Activates PD-1 Expression through Activation of the STAT1-IRF2 Pathway in Myeloid Cells. Cells 2024; 13:1163. [PMID: 38995014 PMCID: PMC11240780 DOI: 10.3390/cells13131163] [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: 04/30/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
PD-1 (Programmed cell death protein 1) regulates the metabolic reprogramming of myeloid-derived suppressor cells and myeloid cell differentiation, as well as the type I interferon (IFN-I) signaling pathway in myeloid cells in the tumor microenvironment. PD-1, therefore, is a key inhibitory receptor in myeloid cells. However, the regulation of PD-1 expression in myeloid cells is unknown. We report that the expression level of PDCD1, the gene that encodes the PD-1 protein, is positively correlated with the levels of IFNB1 and IFNAR1 in myeloid cells in human colorectal cancer. Treatment of mouse myeloid cell lines with recombinant IFNβ protein elevated PD-1 expression in myeloid cells in vitro. Knocking out IFNAR1, the gene that encodes the IFN-I-specific receptor, diminished the inductive effect of IFNβ on PD-1 expression in myeloid cells in vitro. Treatment of tumor-bearing mice with a lipid nanoparticle-encapsulated IFNβ-encoding plasmid (IFNBCOL01) increased IFNβ expression, resulting in elevated PD-1 expression in tumor-infiltrating myeloid cells. At the molecular level, we determined that IFNβ activates STAT1 (signal transducer and activator of transcription 1) and IRFs (interferon regulatory factors) in myeloid cells. Analysis of the cd279 promoter identified IRF2-binding consensus sequence elements. ChIP (chromatin immunoprecipitation) analysis determined that the pSTAT1 directly binds to the irf2 promoter and that IRF2 directly binds to the cd279 promoter in myeloid cells in vitro and in vivo. In colon cancer patients, the expression levels of STAT1, IRF2 and PDCD1 are positively correlated in tumor-infiltrating myeloid cells. Our findings determine that IFNβ activates PD-1 expression at least in part by an autocrine mechanism via the stimulation of the pSTAT1-IRF2 axis in myeloid cells.
Collapse
Affiliation(s)
- Liyan Liang
- School of Life Sciences, Tianjin University, Tianjin 300072, China; (L.L.); (Y.Y.); (K.D.); (Y.W.); (Y.L.)
| | - Yingcui Yang
- School of Life Sciences, Tianjin University, Tianjin 300072, China; (L.L.); (Y.Y.); (K.D.); (Y.W.); (Y.L.)
| | - Kaidi Deng
- School of Life Sciences, Tianjin University, Tianjin 300072, China; (L.L.); (Y.Y.); (K.D.); (Y.W.); (Y.L.)
| | - Yanmin Wu
- School of Life Sciences, Tianjin University, Tianjin 300072, China; (L.L.); (Y.Y.); (K.D.); (Y.W.); (Y.L.)
| | - Yan Li
- School of Life Sciences, Tianjin University, Tianjin 300072, China; (L.L.); (Y.Y.); (K.D.); (Y.W.); (Y.L.)
| | - Liya Bai
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; (L.B.); (Y.W.)
| | - Yinsong Wang
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; (L.B.); (Y.W.)
| | - Chunwan Lu
- School of Life Sciences, Tianjin University, Tianjin 300072, China; (L.L.); (Y.Y.); (K.D.); (Y.W.); (Y.L.)
| |
Collapse
|
7
|
Cadiou G, Beauvais T, Marotte L, Lambot S, Deleine C, Vignes C, Gantier M, Hussong M, Rulli S, Jarry A, Simon S, Malissen B, Labarriere N. Differential impact of genetic deletion of TIGIT or PD-1 on melanoma-specific T-lymphocytes. Oncoimmunology 2024; 13:2376782. [PMID: 38983599 PMCID: PMC11232637 DOI: 10.1080/2162402x.2024.2376782] [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: 05/23/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of PDCD1 or TIGIT genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1KO T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGITKO T-cells. Functional analyses showed that PD-1KO and TIGITKO T-cells displayed superior antitumor reactivity than their wild-type counterpart in vitro and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGITKO T-cells were more effective at inhibiting tumor cell proliferation in vivo, and persist longer within tumors than PD-1KO T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Gwenann Cadiou
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Tiffany Beauvais
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Lucine Marotte
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Sylvia Lambot
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Cécile Deleine
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Caroline Vignes
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Malika Gantier
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
| | - Melanie Hussong
- QIAGEN Sciences, Frederick, MD, USA
- NeoGenomics, Research Triangle Park, Durham, NC, USA
| | | | - Anne Jarry
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Sylvain Simon
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bernard Malissen
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Nathalie Labarriere
- Immunology and New Concepts in ImmunoTherapy, INCIT, Nantes Université, Univ Angers, Inserm, Nantes, France
| |
Collapse
|
8
|
Pollard JM, Hynes G, Yin D, Mandal M, Gounari F, Alegre ML, Chong AS. Pregnancy dedifferentiates memory CD8+ T cells into hypofunctional cells with exhaustion-enriched programs. JCI Insight 2024; 9:e176381. [PMID: 38771643 PMCID: PMC11383355 DOI: 10.1172/jci.insight.176381] [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: 10/03/2023] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
Alloreactive memory, unlike naive, CD8+ T cells resist transplantation tolerance protocols and are a critical barrier to long-term graft acceptance in the clinic. We here show that semiallogeneic pregnancy successfully reprogrammed memory fetus/graft-specific CD8+ T cells (TFGS) toward hypofunction. Female C57BL/6 mice harboring memory CD8+ T cells generated by the rejection of BALB/c skin grafts and then mated with BALB/c males achieved rates of pregnancy comparable with naive controls. Postpartum CD8+ TFGS from skin-sensitized dams upregulated expression of T cell exhaustion (TEX) markers (Tox, Eomes, PD-1, TIGIT, and Lag3). Transcriptional analysis corroborated an enrichment of canonical TEX genes in postpartum memory TFGS and revealed a downregulation of a subset of memory-associated transcripts. Strikingly, pregnancy induced extensive epigenetic modifications of exhaustion- and memory-associated genes in memory TFGS, whereas minimal epigenetic modifications were observed in naive TFGS. Finally, postpartum memory TFGS durably expressed the exhaustion-enriched phenotype, and their susceptibility to transplantation tolerance was significantly restored compared with memory TFGS. These findings advance the concept of pregnancy as an epigenetic modulator inducing hypofunction in memory CD8+ T cells that has relevance not only for pregnancy and transplantation tolerance, but also for tumor immunity and chronic infections.
Collapse
Affiliation(s)
| | - Grace Hynes
- Section of Transplantation, Department of Surgery, and
| | - Dengping Yin
- Section of Transplantation, Department of Surgery, and
| | - Malay Mandal
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Fotini Gounari
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Department of Immunology, Mayo Clinic, Phoenix, Arizona, USA
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Anita S Chong
- Section of Transplantation, Department of Surgery, and
| |
Collapse
|
9
|
Gao M, Shi J, Xiao X, Yao Y, Chen X, Wang B, Zhang J. PD-1 regulation in immune homeostasis and immunotherapy. Cancer Lett 2024; 588:216726. [PMID: 38401888 DOI: 10.1016/j.canlet.2024.216726] [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: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/10/2024] [Indexed: 02/26/2024]
Abstract
Harnessing the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis is pivotal in autoimmunity and cancer immunotherapy. PD-1 receptors on immune cells engage with one of its ligands, PD-L1 or PD-L2, expressed on antigen-presenting cells or tumor cells, driving T-cell dysfunction and tumor immune escape. Thus, targeting PD-1/PD-L1 revitalizes cytotoxic T cells for cancer elimination. However, a majority of cancer patients don't respond to PD-1/PD-L1 blockade, and the underlying mechanisms remain partially understood. Recent studies have revealed that PD-1 expression levels or modifications impact the effectiveness of anti-PD-1/PD-L1 treatments. Therefore, understanding the molecular mechanisms governing PD-1 expression and modifications is crucial for innovating therapeutic strategies to enhance the efficacy of PD-1/PD-L1 inhibition. This article presents a comprehensive overview of advancements in PD-1 regulation and highlights their potential in modulating immune homeostasis and cancer immunotherapy, aiming to refine clinical outcomes.
Collapse
Affiliation(s)
- Minling Gao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jie Shi
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiangling Xiao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yingmeng Yao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xu Chen
- Chongqing University Medical School, Chongqing, 400044, China
| | - Bin Wang
- Department of Gastroenterology & Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Jinfang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
10
|
Petroni G, Pillozzi S, Antonuzzo L. Exploiting Tertiary Lymphoid Structures to Stimulate Antitumor Immunity and Improve Immunotherapy Efficacy. Cancer Res 2024; 84:1199-1209. [PMID: 38381540 PMCID: PMC11016894 DOI: 10.1158/0008-5472.can-23-3325] [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: 10/23/2023] [Revised: 01/04/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024]
Abstract
Tumor-associated tertiary lymphoid structures (TLS) have been associated with favorable clinical outcomes and response to immune checkpoint inhibitors in many cancer types, including non-small cell lung cancer. Although the detailed cellular and molecular mechanisms underlying these clinical associations have not been fully elucidated, growing preclinical and clinical studies are helping to elucidate the mechanisms at the basis of TLS formation, composition, and regulation of immune responses. However, a major challenge remains how to exploit TLS to enhance naïve and treatment-mediated antitumor immune responses. Here, we discuss the current understanding of tumor-associated TLS, preclinical models that can be used to study them, and potential therapeutic interventions to boost TLS formation, with a particular focus on lung cancer research.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Firenze, Italy
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
- Clinical Oncology Unit, Careggi University Hospital, Firenze, Italy
| |
Collapse
|
11
|
Yu KQ, Li CF, Ye L, Song Y, Wang YH, Lin YR, Liao ST, Mei ZC, Lv L. Long Non-Coding RNA ANRIL Regulates Inflammatory Factor Expression in Ulcerative Colitis Via the miR-191-5p/SATB1 Axis. Inflammation 2024; 47:513-529. [PMID: 37985573 DOI: 10.1007/s10753-023-01925-z] [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: 09/21/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023]
Abstract
Ulcerative colitis, an inflammatory bowel disease, manifests with symptoms such as abdominal pain, diarrhea, and mucopurulent feces. The long non-coding RNA (lncRNA) ANRIL exhibits significantly reduced expression in UC, yet its specific mechanism is unknown. This study revealed that ANRIL is involved in the progression of UC by inhibiting IL-6 and TNF-α via miR-191-5P/SATB1 axis. We found that in patients with UC, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were significantly overexpressed in inflamed colon sites, whereas ANRIL was significantly under-expressed and associated with disease severity. The downregulation of ANRIL resulted in the increased expression of IL-6 and TNF-α in LPS-treated FHCs. ANRIL directly targeted miR-191-5p, thereby inhibiting its expression and augmenting SATB1 expression. Moreover, overexpression of miR-191-5p abolished ANRIL-mediated inhibition of IL-6 and TNF-α production. Dual luciferase reporter assays revealed the specific binding of miR-191-5p to ANRIL and SATB1. Furthermore, the downregulation of ANRIL promoted DSS-induced colitis in mice. Together, we provide evidence that ANRIL plays a critical role in regulating IL-6 and TNF-α expression in UC by modulating the miR-191-5p/SATB1 axis. Our study provides novel insights into progression and molecular therapeutic strategies in UC.
Collapse
Affiliation(s)
- Ke-Qi Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Chuan-Fei Li
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Lu Ye
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Ya Song
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Yan-Hui Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Yu-Ru Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China
| | - Sheng-Tao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China.
| | - Zhe-Chuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China.
| | - Lin Lv
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong, Chongqing, 400010, China.
| |
Collapse
|
12
|
Cerchietti L. Genetic mechanisms underlying tumor microenvironment composition and function in diffuse large B-cell lymphoma. Blood 2024; 143:1101-1111. [PMID: 38211334 PMCID: PMC10972714 DOI: 10.1182/blood.2023021002] [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: 10/11/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
ABSTRACT Cells in the tumor microenvironment (TME) of diffuse large B-cell lymphoma (DLBCL) show enormous diversity and plasticity, with functions that can range from tumor inhibitory to tumor supportive. The patient's age, immune status, and DLBCL treatments are factors that contribute to the shaping of this TME, but evidence suggests that genetic factors, arising principally in lymphoma cells themselves, are among the most important. Here, we review the current understanding of the role of these genetic drivers of DLBCL in establishing and modulating the lymphoma microenvironment. A better comprehension of the relationship between lymphoma genetic factors and TME biology should lead to better therapeutic interventions, especially immunotherapies.
Collapse
Affiliation(s)
- Leandro Cerchietti
- Hematology and Oncology Division, Medicine Department, New York-Presbyterian Hospital, Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, NY
| |
Collapse
|
13
|
Yang F, Akhtar MN, Zhang D, El-Mayta R, Shin J, Dorsey JF, Zhang L, Xu X, Guo W, Bagley SJ, Fuchs SY, Koumenis C, Lathia JD, Mitchell MJ, Gong Y, Fan Y. An immunosuppressive vascular niche drives macrophage polarization and immunotherapy resistance in glioblastoma. SCIENCE ADVANCES 2024; 10:eadj4678. [PMID: 38416830 PMCID: PMC10901371 DOI: 10.1126/sciadv.adj4678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Cancer immunity is subjected to spatiotemporal regulation by leukocyte interaction with neoplastic and stromal cells, contributing to immune evasion and immunotherapy resistance. Here, we identify a distinct mesenchymal-like population of endothelial cells (ECs) that form an immunosuppressive vascular niche in glioblastoma (GBM). We reveal a spatially restricted, Twist1/SATB1-mediated sequential transcriptional activation mechanism, through which tumor ECs produce osteopontin to promote immunosuppressive macrophage (Mφ) phenotypes. Genetic or pharmacological ablation of Twist1 reverses Mφ-mediated immunosuppression and enhances T cell infiltration and activation, leading to reduced GBM growth and extended mouse survival, and sensitizing tumor to chimeric antigen receptor T immunotherapy. Thus, these findings uncover a spatially restricted mechanism controlling tumor immunity and suggest that targeting endothelial Twist1 may offer attractive opportunities for optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Junyoung Shin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jay F. Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin D. Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Maymí VI, Zhu H, Jager M, Johnson S, Getchell R, Casey JW, Grenier JK, Wherry EJ, Smith NL, Grimson A, Rudd BD. Neonatal CD8+ T Cells Resist Exhaustion during Chronic Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:834-843. [PMID: 38231127 PMCID: PMC11298781 DOI: 10.4049/jimmunol.2300396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
Chronic viral infections, such as HIV and hepatitis C virus, represent a major public health problem. Although it is well understood that neonates and adults respond differently to chronic viral infections, the underlying mechanisms remain unknown. In this study, we transferred neonatal and adult CD8+ T cells into a mouse model of chronic infection (lymphocytic choriomeningitis virus clone 13) and dissected out the key cell-intrinsic differences that alter their ability to protect the host. Interestingly, we found that neonatal CD8+ T cells preferentially became effector cells early in chronic infection compared with adult CD8+ T cells and expressed higher levels of genes associated with cell migration and effector cell differentiation. During the chronic phase of infection, the neonatal cells retained more immune functionality and expressed lower levels of surface markers and genes related to exhaustion. Because the neonatal cells protect from viral replication early in chronic infection, the altered differentiation trajectories of neonatal and adult CD8+ T cells is functionally significant. Together, our work demonstrates how cell-intrinsic differences between neonatal and adult CD8+ T cells influence key cell fate decisions during chronic infection.
Collapse
Affiliation(s)
- Viviana I. Maymí
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Hongya Zhu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Mason Jager
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Shawn Johnson
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Rodman Getchell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - James W. Casey
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Jennifer K. Grenier
- Transcriptional Regulation and Expression Facility, Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - E. John Wherry
- Institute for Immunology and Immune Health, and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
15
|
Mecca M, Picerno S, Cortellino S. The Killer's Web: Interconnection between Inflammation, Epigenetics and Nutrition in Cancer. Int J Mol Sci 2024; 25:2750. [PMID: 38473997 DOI: 10.3390/ijms25052750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Inflammation is a key contributor to both the initiation and progression of tumors, and it can be triggered by genetic instability within tumors, as well as by lifestyle and dietary factors. The inflammatory response plays a critical role in the genetic and epigenetic reprogramming of tumor cells, as well as in the cells that comprise the tumor microenvironment. Cells in the microenvironment acquire a phenotype that promotes immune evasion, progression, and metastasis. We will review the mechanisms and pathways involved in the interaction between tumors, inflammation, and nutrition, the limitations of current therapies, and discuss potential future therapeutic approaches.
Collapse
Affiliation(s)
- Marisabel Mecca
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Simona Picerno
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Salvatore Cortellino
- Laboratory of Preclinical and Translational Research, Responsible Research Hospital, 86100 Campobasso, CB, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, 80138 Naples, NA, Italy
- S.H.R.O. Italia Foundation ETS, 10060 Candiolo, TO, Italy
| |
Collapse
|
16
|
Falkowski L, Buddenkotte J, Datsi A. Epigenetics in T-cell driven inflammation and cancer. Semin Cell Dev Biol 2024; 154:250-260. [PMID: 36641367 DOI: 10.1016/j.semcdb.2023.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/15/2023]
Abstract
For decades, scientists have been investigating how processes such as gene expression, stem cell plasticity, and cell differentiation can be modulated. The discovery of epigenetics helped unravel these processes and enabled the identification of major underlying mechanisms that, for example, are central for T cell maturation. T cells go through various stages in their development evolving from progenitor cells into double positive CD4/CD8 T cells that finally leave the thymus as naïve T cells. One major mechanism driving T cell maturation is the modulation of gene activity by temporally sequenced transcription of spatially exposed gene loci. DNA methylation, demethylation, and acetylation are key processes that enable a sequenced gene expression required for T cell differentiation. In vivo, differentiated T cells are subjected to enormous pressures originating from the microenvironment. Signals from this environment, particularly from an inflammatory or a tumor microenvironment, can push T cells to differentiate into specific effector and memory T cells, and even prompt T cells to adopt a state of dysfunctional exhaustion, en route of an epigenetically controlled mechanism. Fundamentals of these processes will be discussed in this review highlighting potential therapeutic interventions, in particular those beneficial to revive exhausted T cells.
Collapse
Affiliation(s)
- Lea Falkowski
- Institute for Transplantational Diagnostics and Cell Therapeutics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Joerg Buddenkotte
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Angeliki Datsi
- Institute for Transplantational Diagnostics and Cell Therapeutics, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
17
|
Kolahi Azar H, Imanpour A, Rezaee H, Ezzatifar F, Zarei-Behjani Z, Rostami M, Azami M, Behestizadeh N, Rezaei N. Mesenchymal stromal cells and CAR-T cells in regenerative medicine: The homing procedure and their effective parameters. Eur J Haematol 2024; 112:153-173. [PMID: 37254607 DOI: 10.1111/ejh.14014] [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: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Mesenchymal stromal cells (MSCs) and chimeric antigen receptor (CAR)-T cells are two core elements in cell therapy procedures. MSCs have significant immunomodulatory effects that alleviate inflammation in the tissue regeneration process, while administration of specific chemokines and adhesive molecules would primarily facilitate CAR-T cell trafficking into solid tumors. Multiple parameters affect cell homing, including the recipient's age, the number of cell passages, proper cell culture, and the delivery method. In addition, several chemokines are involved in the tumor microenvironment, affecting the homing procedure. This review discusses parameters that improve the efficiency of cell homing and significant cell therapy challenges. Emerging comprehensive mechanistic strategies such as non-systemic and systemic homing that revealed a significant role in cell therapy remodeling were also reviewed. Finally, the primary implications for the development of combination therapies that incorporate both MSCs and CAR-T cells for cancer treatment were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aylar Imanpour
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh Rezaee
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ezzatifar
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeinab Zarei-Behjani
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, Advanced School of Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Behestizadeh
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
18
|
Liao X, Li W, Zhou H, Rajendran BK, Li A, Ren J, Luan Y, Calderwood DA, Turk B, Tang W, Liu Y, Wu D. The CUL5 E3 ligase complex negatively regulates central signaling pathways in CD8 + T cells. Nat Commun 2024; 15:603. [PMID: 38242867 PMCID: PMC10798966 DOI: 10.1038/s41467-024-44885-0] [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: 10/21/2022] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
CD8+ T cells play an important role in anti-tumor immunity. Better understanding of their regulation could advance cancer immunotherapies. Here we identify, via stepwise CRISPR-based screening, that CUL5 is a negative regulator of the core signaling pathways of CD8+ T cells. Knocking out CUL5 in mouse CD8+ T cells significantly improves their tumor growth inhibiting ability, with significant proteomic alterations that broadly enhance TCR and cytokine signaling and their effector functions. Chemical inhibition of neddylation required by CUL5 activation, also enhances CD8 effector activities with CUL5 validated as a major target. Mechanistically, CUL5, which is upregulated by TCR stimulation, interacts with the SOCS-box-containing protein PCMTD2 and inhibits TCR and IL2 signaling. Additionally, CTLA4 is markedly upregulated by CUL5 knockout, and its inactivation further enhances the anti-tumor effect of CUL5 KO. These results together reveal a negative regulatory mechanism for CD8+ T cells and have strong translational implications in cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaofeng Liao
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Wenxue Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Hongyue Zhou
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Barani Kumar Rajendran
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Ao Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jingjing Ren
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yi Luan
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - David A Calderwood
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Benjamin Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Wenwen Tang
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Yale Cancer Research Institute, Yale University School of Medicine, West Haven, CT, 06516, USA.
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Dianqing Wu
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
19
|
Chung H, Gyu-mi P, Na YR, Lee YS, Choi H, Seok SH. Comprehensive characterization of early-programmed tumor microenvironment by tumor-associated macrophages reveals galectin-1 as an immune modulatory target in breast cancer. Theranostics 2024; 14:843-860. [PMID: 38169569 PMCID: PMC10758049 DOI: 10.7150/thno.88917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Background: In recent years, there has been considerable interest in the therapeutic targeting of tumor-associated macrophages (TAMs) to modulate the tumor microenvironment (TME), resulting in antitumoral phenotypes. However, key mediators suitable for TAM-mediated remodeling of the TME remain poorly understood. Methods: In this study, we used single-cell RNA sequencing analyses to analyze the landscape of the TME modulated by TAMs in terms of a protumor microenvironment during early tumor development. Results: Our data revealed that the depletion of TAMs leads to a decreased epithelial-to-mesenchymal transition (EMT) signature in cancer cells and a distinct transcriptional state characterized by CD8+ T cell activation. Moreover, notable alterations in gene expression were observed upon the depletion of TAMs, identifying Galectin-1 (Gal-1) as a crucial molecular factor responsible for the observed effect. Gal-1 inhibition reversed immune suppression via the reinvigoration of CD8+ T cells, impairing tumor growth and potentiating immune checkpoint inhibitors in breast tumor models. Conclusion: These results provide comprehensive insights into TAM-mediated early tumor microenvironments and reveal immune evasion mechanisms that can be targeted by Gal-1 to induce antitumor immune responses.
Collapse
Affiliation(s)
- Hyewon Chung
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, Republic of Korea
| | - Park Gyu-mi
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yi Rang Na
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
20
|
Zhu Z, Lou G, Teng XL, Wang H, Luo Y, Shi W, Yihunie K, Hao S, DeGolier K, Liao C, Huang H, Zhang Q, Fry T, Wang T, Yao C, Wu T. FOXP1 and KLF2 reciprocally regulate checkpoints of stem-like to effector transition in CAR T cells. Nat Immunol 2024; 25:117-128. [PMID: 38012417 PMCID: PMC10841689 DOI: 10.1038/s41590-023-01685-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 10/16/2023] [Indexed: 11/29/2023]
Abstract
In cancer and infections, self-renewing stem-like CD8+ T cells mediate the response of immunotherapies and replenish terminally exhausted T cells and effector-like T cells. However, the programs governing the lineage choice in chimeric antigen receptor (CAR) T cells are unclear. Here, by simultaneously profiling single-cell chromatin accessibility and transcriptome in the same CAR T cells, we identified heterogeneous chromatin states within CD8+ T cell subsets that foreshadowed transcriptional changes and were primed for regulation by distinct transcription factors. Transcription factors that controlled each CD8+ T cell subset were regulated by high numbers of enhancers and positioned as hubs of gene networks. FOXP1, a hub in the stem-like network, promoted expansion and stemness of CAR T cells and limited excessive effector differentiation. In the effector network, KLF2 enhanced effector CD8+ T cell differentiation and prevented terminal exhaustion. Thus, we identified gene networks and hub transcription factors that controlled the differentiation of stem-like CD8+ CAR T cells into effector or exhausted CD8+ CAR T cells.
Collapse
Affiliation(s)
- Ziang Zhu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Immunology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guohua Lou
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Xiao-Lu Teng
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haixia Wang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ying Luo
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wangke Shi
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kiddist Yihunie
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shumeng Hao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kole DeGolier
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huocong Huang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Terry Fry
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tao Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
21
|
Bromley JD, Ganchua SKC, Nyquist SK, Maiello P, Chao M, Borish HJ, Rodgers M, Tomko J, Kracinovsky K, Mugahid D, Nguyen S, Wang D, Rosenberg JM, Klein EC, Gideon HP, Floyd-O’Sullivan R, Berger B, Scanga CA, Lin PL, Fortune SM, Shalek AK, Flynn JL. CD4 + T cells are homeostatic regulators during Mtb reinfection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572669. [PMID: 38187598 PMCID: PMC10769325 DOI: 10.1101/2023.12.20.572669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Immunological priming - either in the context of prior infection or vaccination - elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Here, using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrate that prior Mtb infection elicits a long-lasting protective response against subsequent Mtb exposure and that the depletion of CD4+ T cells prior to Mtb rechallenge significantly abrogates this protection. Leveraging microbiologic, PET-CT, flow cytometric, and single-cell RNA-seq data from primary infection, reinfection, and reinfection-CD4+ T cell depleted granulomas, we identify differential cellular and microbial features of control. The data collectively demonstrate that the presence of CD4+ T cells in the setting of reinfection results in a reduced inflammatory lung milieu characterized by reprogrammed CD8+ T cell activity, reduced neutrophilia, and blunted type-1 immune signaling among myeloid cells, mitigating Mtb disease severity. These results open avenues for developing vaccines and therapeutics that not only target CD4+ and CD8+ T cells, but also modulate innate immune cells to limit Mtb disease.
Collapse
Affiliation(s)
- Joshua D. Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharie Keanne C. Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Sarah K. Nyquist
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
| | - Michael Chao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Mark Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Kara Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Douaa Mugahid
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Son Nguyen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Wang
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob M. Rosenberg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edwin C. Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah P. Gideon
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Roisin Floyd-O’Sullivan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine
| | - Sarah M. Fortune
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alex K. Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
- Lead contact
| |
Collapse
|
22
|
Hao L, Li S, Hu X. New insights into T-cell exhaustion in liver cancer: from mechanism to therapy. J Cancer Res Clin Oncol 2023; 149:12543-12560. [PMID: 37423958 DOI: 10.1007/s00432-023-05083-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
Liver cancer is one of the most common malignancies. T-cell exhaustion is associated with immunosuppression of tumor and chronic infection. Although immunotherapies that enhance the immune response by targeting programmed cell death-1(PD-1)/programmed cell death ligand 1 (PD-L1) have been applied to malignancies, these treatments have shown limited response rates. This suggested that additional inhibitory receptors (IRs) also contributed to T-cell exhaustion and tumor prognosis. Exhausted T-cells (Tex) in the tumor immune microenvironment (TME) are usually in a dysfunctional state of exhaustion, such as impaired activity and proliferative ability, increased apoptosis rate, and reduced production of effector cytokines. Tex cells participate in the negative regulation of tumor immunity mainly through IRs on the cell surface, changes in cytokines and immunomodulatory cell types, causing tumor immune escape. However, T-cell exhaustion is not irreversible and targeted immune checkpoint inhibitors (ICIs) can effectively reverse the exhaustion of T-cells and restore the anti-tumor immune response. Therefore, the research on the mechanism of T-cell exhaustion in liver cancer, aimed at maintaining or restoring the effector function of Tex cells, might provide a new method for the treatment of liver cancer. In this review, we summarized the basic characteristics of Tex cells (such as IRs and cytokines), discussed the mechanisms associated with T-cell exhaustion, and specifically discussed how these exhaustion characteristics were acquired and shaped by key factors within TME. Then new insights into the molecular mechanism of T-cell exhaustion suggested a potential way to improve the efficacy of cancer immunotherapy, namely to restore the effector function of Tex cells. In addition, we also reviewed the research progress of T-cell exhaustion in recent years and provided suggestions for further research.
Collapse
Affiliation(s)
- Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-Er-Qiao Road, Chengdu, 610075, Sichuan Province, People's Republic of China
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-Er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
23
|
Harro CM, Sprenger KB, Chaurio RA, Powers JJ, Innamarato P, Anadon CM, Zhang Y, Biswas S, Mandal G, Mine JA, Cortina C, Nagy MZ, Martin AL, Handley KF, Borjas GJ, Chen PL, Pinilla-Ibarz J, Sokol L, Yu X, Conejo-Garcia JR. Sézary syndrome originates from heavily mutated hematopoietic progenitors. Blood Adv 2023; 7:5586-5602. [PMID: 37531660 PMCID: PMC10514084 DOI: 10.1182/bloodadvances.2022008562] [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: 07/14/2022] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/04/2023] Open
Abstract
The pathogenesis of cutaneous T-cell lymphoma (CTCL) remains unclear. Using single-cell RNA or T-cell receptor (TCR) sequencing of 32 619 CD3+CD4+ and CD26+/CD7+ and 29 932 CD3+CD4+ and CD26-/CD7- lymphocytes from the peripheral blood of 7 patients with CTCL, coupled to single-cell ATAC-sequencing of 26,411 CD3+CD4+ and CD26+/CD7+ and 33 841 CD3+CD4+ and CD26-/CD7- lymphocytes, we show that tumor cells in Sézary syndrome and mycosis fungoides (MF) exhibit different phenotypes and trajectories of differentiation. When compared to MF, Sézary cells exhibit narrower repertoires of TCRs and exhibit clonal enrichment. Surprisingly, we identified ≥200 mutations in hematopoietic stem cells from multiple patients with Sézary syndrome. Mutations in key oncogenes were also present in peripheral Sézary cells, which also showed the hallmarks of recent thymic egression. Together our data suggest that CTCL arises from mutated lymphocyte progenitors that acquire TCRs in the thymus, which complete their malignant transformation in the periphery.
Collapse
Affiliation(s)
- Carly M. Harro
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL
- Cancer Biology PhD Program, College of Arts and Sciences, University of South Florida, Tampa, FL
| | - Kimberly B. Sprenger
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Ricardo A. Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Immunology, Duke School of Medicine, Durham, NC
| | - John J. Powers
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Patrick Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Carmen M. Anadon
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Immunology, Duke School of Medicine, Durham, NC
| | - Yumeng Zhang
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Gunjan Mandal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Biotechnology, Institute of Life Sciences, Bhubaneswar, India
| | - Jessica A. Mine
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Immunology, Duke School of Medicine, Durham, NC
| | - Carla Cortina
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Mate Z. Nagy
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Alexandra L. Martin
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Katelyn F. Handley
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Gustavo J. Borjas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Pei-Ling Chen
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Javier Pinilla-Ibarz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Lubomir Sokol
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Jose R. Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Immunology, Duke School of Medicine, Durham, NC
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
24
|
Lan X, Zebley CC, Youngblood B. Cellular and molecular waypoints along the path of T cell exhaustion. Sci Immunol 2023; 8:eadg3868. [PMID: 37656775 PMCID: PMC10618911 DOI: 10.1126/sciimmunol.adg3868] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/09/2023] [Indexed: 09/03/2023]
Abstract
Thirty years of foundational research investigating molecular and cellular mechanisms promoting T cell exhaustion are now enabling rational design of T cell-based therapies for the treatment of chronic infections and cancer. Once described as a static cell fate, it is now well appreciated that the developmental path toward exhaustion is composed of a heterogeneous pool of cells with varying degrees of effector potential that ultimately converge on a terminally differentiated state. Recent description of the developmental stages along the differentiation trajectory of T cell exhaustion has provided insight into past immunotherapeutic success and future opportunities. Here, we discuss the hallmarks of distinct developmental stages occurring along the path to T cell dysfunction and the impact of these discrete CD8+ T cell fates on cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Lan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Caitlin C. Zebley
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ben Youngblood
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
25
|
Nomura A, Kobayashi T, Seo W, Ohno-Oishi M, Kakugawa K, Muroi S, Yoshida H, Endo TA, Moro K, Taniuchi I. Identification of a novel enhancer essential for Satb1 expression in T H2 cells and activated ILC2s. Life Sci Alliance 2023; 6:e202301897. [PMID: 37193606 PMCID: PMC10189277 DOI: 10.26508/lsa.202301897] [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: 01/03/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023] Open
Abstract
The genome organizer, special AT-rich binding protein-1 (SATB1), functions to globally regulate gene networks during primary T cell development and plays a pivotal role in lineage specification in CD4+ helper-, CD8+ cytotoxic-, and FOXP3+ regulatory-T cell subsets. However, it remains unclear how Satb1 gene expression is controlled, particularly in effector T cell function. Here, by using a novel reporter mouse strain expressing SATB1-Venus and genome editing, we have identified a cis-regulatory enhancer, essential for maintaining Satb1 expression specifically in TH2 cells. This enhancer is occupied by STAT6 and interacts with Satb1 promoters through chromatin looping in TH2 cells. Reduction of Satb1 expression, by the lack of this enhancer, resulted in elevated IL-5 expression in TH2 cells. In addition, we found that Satb1 is induced in activated group 2 innate lymphoid cells (ILC2s) through this enhancer. Collectively, these results provide novel insights into how Satb1 expression is regulated in TH2 cells and ILC2s during type 2 immune responses.
Collapse
Affiliation(s)
- Aneela Nomura
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Tetsuro Kobayashi
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Wooseok Seo
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Michiko Ohno-Oishi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Kiyokazu Kakugawa
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Sawako Muroi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Hideyuki Yoshida
- Laboratory for YCI Laboratory for Immunological Transcriptomics, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Takaho A Endo
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
- Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School for Medicine, Osaka University, Osaka, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
| |
Collapse
|
26
|
Kumari R, Hosseini ES, Warrington KE, Milonas T, Payne KK. Butyrophilins: Dynamic Regulators of Protective T Cell Immunity in Cancer. Int J Mol Sci 2023; 24:8722. [PMID: 37240071 PMCID: PMC10218201 DOI: 10.3390/ijms24108722] [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: 04/13/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The efficacy of current immunotherapies remains limited in many solid epithelial malignancies. Recent investigations into the biology of butyrophilin (BTN) and butyrophilin-like (BTNL) molecules, however, suggest these molecules are potent immunosuppressors of antigen-specific protective T cell activity in tumor beds. BTN and BTNL molecules also associate with each other dynamically on cellular surfaces in specific contexts, which modulates their biology. At least in the case of BTN3A1, this dynamism drives the immunosuppression of αβ T cells or the activation of Vγ9Vδ2 T cells. Clearly, there is much to learn regarding the biology of BTN and BTNL molecules in the context of cancer, where they may represent intriguing immunotherapeutic targets that could potentially synergize with the current class of immune modulators in cancer. Here, we discuss our current understanding of BTN and BTNL biology, with a particular focus on BTN3A1, and potential therapeutic implications for cancer.
Collapse
Affiliation(s)
- Rinkee Kumari
- Medical Immunology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Elaheh Sadat Hosseini
- Medical Immunology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Cellular and Molecular Pharmacology, Rutgers School of Graduate Studies, Rutgers, The State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Kristen E. Warrington
- Medical Immunology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Tyler Milonas
- Medical Immunology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Kyle K. Payne
- Medical Immunology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Cellular and Molecular Pharmacology, Rutgers School of Graduate Studies, Rutgers, The State University of New Jersey, New Brunswick, NJ 08854, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
27
|
Ford BR, Poholek AC. Regulation and Immunotherapeutic Targeting of the Epigenome in Exhausted CD8 T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:869-879. [PMID: 36947818 PMCID: PMC10037537 DOI: 10.4049/jimmunol.2200681] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/04/2023] [Indexed: 03/24/2023]
Abstract
Exhaustion is a state of CD8 T cell differentiation that occurs in settings of chronic Ag such as tumors, chronic viral infection, and autoimmunity. Cellular differentiation is driven by a series of environmental signals that promote epigenetic landscapes that set transcriptomes needed for function. For CD8 T cells, the epigenome that underlies exhaustion is distinct from effector and memory cell differentiation, suggesting that signals early on set in motion a process where the epigenome is modified to promote a trajectory toward a dysfunctional state. Although we know many signals that promote exhaustion, putting this in the context of the epigenetic changes that occur during differentiation has been less clear. In this review, we aim to summarize the epigenetic changes associated with exhaustion in the context of signals that promote it, highlighting immunotherapeutic studies that support these observations or areas for future therapeutic opportunities.
Collapse
Affiliation(s)
- B Rhodes Ford
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA; and Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Amanda C Poholek
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA; and Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
28
|
Transcriptomic profiling of programmed cell death 1 (PD-1) expressing T cells in early rheumatoid arthritis identifies a decreased CD4 + PD-1 + signature post-treatment. Sci Rep 2023; 13:2847. [PMID: 36801909 PMCID: PMC9938264 DOI: 10.1038/s41598-023-29971-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Programmed cell death protein 1 (PD-1)-expressing T cells are expanded in individuals with established rheumatoid arthritis (RA). However, little is known about their functional role in the pathogenesis of early RA. To address this, we investigated the transcriptomic profiles of circulating CD4+ and CD8+ PD-1+ lymphocytes from patients with early RA (n = 5) using fluorescence activated cell sorting in conjunction with total RNA sequencing. Additionally, we assessed for alterations in CD4+PD-1+ gene signatures in previously published synovial tissue (ST) biopsy data (n = 19) (GSE89408, GSE97165) before and after six-months of triple disease modifying anti-rheumatic drug (tDMARD) treatment. Comparisons of gene signatures between CD4+PD-1+ vs. PD-1- cells identified significant upregulation of genes including CXCL13 and MAF, and in pathways including Th1 and Th2, cross talk between dendritic cells and NK cells, B cell development and antigen presentation. Gene signatures from early RA ST before and after six-month tDMARD treatment revealed downregulation of the CD4+PD-1+ signatures following treatment, identifying a mechanism through which tDMARDs exert their effect by influencing T cell populations. Furthermore, we identify factors associated with B cell help that are enhanced in the ST compared with PBMCs, highlighting their importance in driving synovial inflammation.
Collapse
|
29
|
Salmond RJ. Regulation of T Cell Activation and Metabolism by Transforming Growth Factor-Beta. BIOLOGY 2023; 12:297. [PMID: 36829573 PMCID: PMC9953227 DOI: 10.3390/biology12020297] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023]
Abstract
Transforming growth factor beta (TGFβ) receptor signalling regulates T cell development, differentiation and effector function. Expression of the immune-associated isoform of this cytokine, TGFβ1, is absolutely required for the maintenance of immunological tolerance in both mice and humans, whilst context-dependent TGFβ1 signalling regulates the differentiation of both anti- and pro-inflammatory T cell effector populations. Thus, distinct TGFβ-dependent T cell responses are implicated in the suppression or initiation of inflammatory and autoimmune diseases. In cancer settings, TGFβ signals contribute to the blockade of anti-tumour immune responses and disease progression. Given the key functions of TGFβ in the regulation of immune responses and the potential for therapeutic targeting of TGFβ-dependent pathways, the mechanisms underpinning these pleiotropic effects have been the subject of much investigation. This review focuses on accumulating evidence suggesting that modulation of T cell metabolism represents a major mechanism by which TGFβ influences T cell immunity.
Collapse
Affiliation(s)
- Robert J Salmond
- Leeds Institute of Medical Research at St. James's, School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
30
|
Lv J, Qin L, Zhao R, Wu D, Wu Z, Zheng D, Li S, Luo M, Wu Q, Long Y, Tang Z, Tang YL, Luo X, Yao Y, Yang LH, Li P. Disruption of CISH promotes the antitumor activity of human T cells and decreases PD-1 expression levels. Mol Ther Oncolytics 2022; 28:46-58. [PMID: 36654786 PMCID: PMC9827364 DOI: 10.1016/j.omto.2022.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor cells and the immunosuppressive tumor microenvironment suppress the antitumor activity of T cells through immune checkpoints, including the PD-L1/PD-1 axis. Cytokine-inducible SH2-containing protein (CISH), a member of the suppressor of cytokine signaling (SOCS) family, inhibits JAK-STAT and T cell receptor (TCR) signaling in T and natural killer (NK) cells. However, its role in the regulation of immune checkpoints in T cells remains unclear. In this study, we ablated CISH in T cells with CRISPR-Cas9 and found that the sensitivity of T cells to TCR and cytokine stimulation was increased. In addition, chimeric antigen receptor T cells with CISH deficiency exhibited longer survival and higher cytokine secretion and antitumor activity. Notably, PD-1 expression was decreased in activated CISH-deficient T cells in vitro and in vivo. The level of FBXO38, a ubiquitination-regulating protein that reduces PD-1 expression, was elevated in activated T cells after CISH ablation. Hence, this study reveals a mechanism by which CISH promotes PD-1 expression by suppressing the expression of FBXO38 and proposes a new strategy for augmenting the therapeutic effect of CAR-T cells by inhibiting CISH.
Collapse
Affiliation(s)
- Jiang Lv
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Le Qin
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ruocong Zhao
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR 999077, China
| | - Di Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhiping Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Diwei Zheng
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Siyu Li
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Mintao Luo
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiting Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Youguo Long
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhaoyang Tang
- Guangdong Zhaotai InVivo Biomedicine Co., Ltd., Guangzhou 510700, China
| | - Yan-Lai Tang
- Department of Paediatrics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xuequn Luo
- Department of Paediatrics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yao Yao
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Li-Hua Yang
- Department of Paediatrics, Zhujiang Hospital, Southern China Medical University, Guangzhou, Guangdong 510280, China,Corresponding author Li-Hua Yang, Department of Paediatrics, Zhujiang Hospital, Southern China Medical University, Guangzhou, Guangdong 510280, China.
| | - Peng Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR 999077, China,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Corresponding author Peng Li, China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
31
|
Cifuentes-Bernal AM, Pham VVH, Li X, Liu L, Li J, Duy Le T. Dynamic cancer drivers: a causal approach for cancer driver discovery based on bio-pathological trajectories. Brief Funct Genomics 2022; 21:455-465. [PMID: 36124841 PMCID: PMC10467634 DOI: 10.1093/bfgp/elac030] [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: 05/18/2022] [Revised: 08/08/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022] Open
Abstract
The traditional way for discovering genes which drive cancer (namely cancer drivers) neglects the dynamic information of cancer development, even though it is well known that cancer progresses dynamically. To enhance cancer driver discovery, we expand cancer driver concept to dynamic cancer driver as a gene driving one or more bio-pathological transitions during cancer progression. Our method refers to the fact that cancer should not be considered as a single process but a compendium of altered biological processes causing the disease to develop over time. Reciprocally, different drivers of cancer can potentially be discovered by analysing different bio-pathological pathways. We propose a novel approach for causal inference of genes driving one or more core processes during cancer development (i.e. dynamic cancer driver). We use the concept of pseudotime for inferring the latent progression of samples along a biological transition during cancer and identifying a critical event when such a process is significantly deviated from normal to carcinogenic. We infer driver genes by assessing the causal effect they have on the process after such a critical event. We have applied our method to single-cell and bulk sequencing datasets of breast cancer. The evaluation results show that our method outperforms well-recognized cancer driver inference methods. These results suggest that including information of the underlying dynamics of cancer improves the inference process (in comparison with using static data), and allows us to discover different sets of driver genes from different processes in cancer. R scripts and datasets can be found at https://github.com/AndresMCB/DynamicCancerDriver.
Collapse
Affiliation(s)
- Andres M Cifuentes-Bernal
- UniSA STEM Unit, University of South Australia,
Mawson Lakes Blvd, 5095, South Australia , Australia
| | - Vu V H Pham
- UniSA STEM Unit, University of South Australia,
Mawson Lakes Blvd, 5095, South Australia , Australia
| | - Xiaomei Li
- UniSA STEM Unit, University of South Australia,
Mawson Lakes Blvd, 5095, South Australia , Australia
| | - Lin Liu
- UniSA STEM Unit, University of South Australia,
Mawson Lakes Blvd, 5095, South Australia , Australia
| | - Jiuyong Li
- UniSA STEM Unit, University of South Australia,
Mawson Lakes Blvd, 5095, South Australia , Australia
| | - Thuc Duy Le
- UniSA STEM Unit, University of South Australia,
Mawson Lakes Blvd, 5095, South Australia , Australia
| |
Collapse
|
32
|
Qin R, Mahal LK, Bojar D. Deep learning explains the biology of branched glycans from single-cell sequencing data. iScience 2022; 25:105163. [PMID: 36217547 PMCID: PMC9547197 DOI: 10.1016/j.isci.2022.105163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/03/2022] Open
Abstract
Glycosylation is ubiquitous and often dysregulated in disease. However, the regulation and functional significance of various types of glycosylation at cellular levels is hard to unravel experimentally. Multi-omics, single-cell measurements such as SUGAR-seq, which quantifies transcriptomes and cell surface glycans, facilitate addressing this issue. Using SUGAR-seq data, we pioneered a deep learning model to predict the glycan phenotypes of cells (mouse T lymphocytes) from transcripts, with the example of predicting β1,6GlcNAc-branching across T cell subtypes (test set F1 score: 0.9351). Model interpretation via SHAP (SHapley Additive exPlanations) identified highly predictive genes, in part known to impact (i) branched glycan levels and (ii) the biology of branched glycans. These genes included physiologically relevant low-abundance genes that were not captured by conventional differential expression analysis. Our work shows that interpretable deep learning models are promising for uncovering novel functions and regulatory mechanisms of glycans from integrated transcriptomic and glycomic datasets.
Collapse
Affiliation(s)
- Rui Qin
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Lara K. Mahal
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Daniel Bojar
- Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
33
|
Gupta PK, Allocco JB, Fraipont JM, McKeague ML, Wang P, Andrade MS, McIntosh C, Chen L, Wang Y, Li Y, Andrade J, Conejo-Garcia JR, Chong AS, Alegre ML. Reduced Satb1 expression predisposes CD4 + T conventional cells to Treg suppression and promotes transplant survival. Proc Natl Acad Sci U S A 2022; 119:e2205062119. [PMID: 36161903 PMCID: PMC9546564 DOI: 10.1073/pnas.2205062119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Limiting CD4+ T cell responses is important to prevent solid organ transplant rejection. In a mouse model of costimulation blockade-dependent cardiac allograft tolerance, we previously reported that alloreactive CD4+ conventional T cells (Tconvs) develop dysfunction, losing proliferative capacity. In parallel, induction of transplantation tolerance is dependent on the presence of regulatory T cells (Tregs). Whether susceptibility of CD4+ Tconvs to Treg suppression is modulated during tolerance induction is unknown. We found that alloreactive Tconvs from transplant tolerant mice had augmented sensitivity to Treg suppression when compared with memory T cells from rejector mice and expressed a transcriptional profile distinct from these memory T cells, including down-regulated expression of the transcription factor Special AT-rich sequence-binding protein 1 (Satb1). Mechanistically, Satb1 deficiency in CD4+ T cells limited their expression of CD25 and IL-2, and addition of Tregs, which express higher levels of CD25 than Satb1-deficient Tconvs and successfully competed for IL-2, resulted in greater suppression of Satb1-deficient than wild-type Tconvs in vitro. In vivo, Satb1-deficient Tconvs were more susceptible to Treg suppression, resulting in significantly prolonged skin allograft survival. Overall, our study reveals that transplantation tolerance is associated with Tconvs' susceptibility to Treg suppression, via modulated expression of Tconv-intrinsic Satb1. Targeting Satb1 in the context of Treg-sparing immunosuppressive therapies might be exploited to improve transplant outcomes.
Collapse
Affiliation(s)
- Pawan K. Gupta
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jennifer B. Allocco
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jane M. Fraipont
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Michelle L. McKeague
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Peter Wang
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Michael S. Andrade
- Section of Transplantation, Department of Surgery, University of Chicago, Chicago, IL 60637
| | - Christine McIntosh
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Luqiu Chen
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Ying Wang
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Yan Li
- Center for Research Informatics, University of Chicago, Chicago, IL 60637
| | - Jorge Andrade
- Center for Research Informatics, University of Chicago, Chicago, IL 60637
| | - José R. Conejo-Garcia
- Department of Immunology, Moffitt Cancer Center & Research Institute, University of South Florida, Tampa, FL 33612
| | - Anita S. Chong
- Section of Transplantation, Department of Surgery, University of Chicago, Chicago, IL 60637
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637
| |
Collapse
|
34
|
Rodriguez-Marquez P, Calleja-Cervantes ME, Serrano G, Oliver-Caldes A, Palacios-Berraquero ML, Martin-Mallo A, Calviño C, Español-Rego M, Ceballos C, Lozano T, San Martin-Uriz P, Vilas-Zornoza A, Rodriguez-Diaz S, Martinez-Turrillas R, Jauregui P, Alignani D, Viguria MC, Redondo M, Pascal M, Martin-Antonio B, Juan M, Urbano-Ispizua A, Rodriguez-Otero P, Alfonso-Pierola A, Paiva B, Lasarte JJ, Inoges S, Lopez-Diaz de Cerio A, San-Miguel J, Fernandez de Larrea C, Hernaez M, Rodriguez-Madoz JR, Prosper F. CAR density influences antitumoral efficacy of BCMA CAR T cells and correlates with clinical outcome. SCIENCE ADVANCES 2022; 8:eabo0514. [PMID: 36179026 PMCID: PMC9524842 DOI: 10.1126/sciadv.abo0514] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 08/17/2022] [Indexed: 05/23/2023]
Abstract
Identification of new markers associated with long-term efficacy in patients treated with CAR T cells is a current medical need, particularly in diseases such as multiple myeloma. In this study, we address the impact of CAR density on the functionality of BCMA CAR T cells. Functional and transcriptional studies demonstrate that CAR T cells with high expression of the CAR construct show an increased tonic signaling with up-regulation of exhaustion markers and increased in vitro cytotoxicity but a decrease in in vivo BM infiltration. Characterization of gene regulatory networks using scRNA-seq identified regulons associated to activation and exhaustion up-regulated in CARHigh T cells, providing mechanistic insights behind differential functionality of these cells. Last, we demonstrate that patients treated with CAR T cell products enriched in CARHigh T cells show a significantly worse clinical response in several hematological malignancies. In summary, our work demonstrates that CAR density plays an important role in CAR T activity with notable impact on clinical response.
Collapse
Affiliation(s)
| | - Maria E. Calleja-Cervantes
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Guillermo Serrano
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Aina Oliver-Caldes
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | | | - Angel Martin-Mallo
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Cristina Calviño
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Marta Español-Rego
- Department of Immunology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Candela Ceballos
- Hematology Service, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Teresa Lozano
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | | | - Amaia Vilas-Zornoza
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Rebeca Martinez-Turrillas
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Patricia Jauregui
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Diego Alignani
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Maria C. Viguria
- Hematology Service, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Margarita Redondo
- Hematology Service, Hospital Universitario de Navarra, IdiSNA, Pamplona, Spain
| | - Mariona Pascal
- Department of Immunology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Beatriz Martin-Antonio
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Manel Juan
- Department of Immunology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
- Immunotherapy platform Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Paula Rodriguez-Otero
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Ana Alfonso-Pierola
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Juan J. Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Susana Inoges
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Immunology and Immunotherapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Ascension Lopez-Diaz de Cerio
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Immunology and Immunotherapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
| | - Jesus San-Miguel
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Carlos Fernandez de Larrea
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Mikel Hernaez
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Data Science and Artificial Intelligence Institute (DATAI), Universidad de Navarra, Pamplona, Spain
| | - Juan R. Rodriguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Felipe Prosper
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| |
Collapse
|
35
|
CMTM3 as a Potential New Immune Checkpoint Regulator. JOURNAL OF ONCOLOGY 2022; 2022:2103515. [PMID: 36245970 PMCID: PMC9553517 DOI: 10.1155/2022/2103515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022]
Abstract
Objectives To evaluate the role of CKLF-like MARVEL transmembrane domain containing 3 (CMTM3) in tumor microenvironment and cancer immunotherapy and explore its potential mechanism. Method The cancer genome map was obtained from the UCSC Xena database. RNAseq data from the Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases were utilized for evaluating the expression and prognostic value of CMTM3 through survival data of clinical trials. The enrichment analysis of CMTM3 was performed using the R package “clusterProfiler.” The scores of immune cell infiltration in TCGA samples were downloaded from the ImmuCellAI database and TIMER2 database, and the relationship between both immune cell invasion and CMTM3 expression was investigated. Immunological activation and suppression genes, immune checkpoints, chemokines, and their receptors were all investigated in relation to CMTM3. Results Most tumor types had varied levels of CMTM3 expression and predicted poor survival status. The CMTM3 expression is closely associated with cancer-associated fibroblasts, macrophages, myeloid dendritic cells, endothelial cells, immune activation genes, immune suppressor genes, immune checkpoints, chemokines, and related receptors. Conclusion Our data reveal that CMTM3 might be used as a cancer biomarker. CMTM3 may work in conjunction with other immunological checkpoints to alter the immune milieu, which could lead to the establishment of new immunotherapy medicines.
Collapse
|
36
|
Nüssing S, Miosge LA, Lee K, Olshansky M, Barugahare A, Roots CM, Sontani Y, Day EB, Koutsakos M, Kedzierska K, Goodnow CC, Russ BE, Daley SR, Turner SJ. SATB1 ensures appropriate transcriptional programs within naïve CD8
+
T cells. Immunol Cell Biol 2022; 100:636-652. [PMID: 35713361 PMCID: PMC9542893 DOI: 10.1111/imcb.12566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 06/07/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Simone Nüssing
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity University of Melbourne Parkville VIC Australia
| | - Lisa A Miosge
- John Curtin School of Medical Research Australian National University Canberra ACT Australia
| | - Kah Lee
- Department of Microbiology, Immunity Theme, Biomedicine Discovery Institute Monash University Clayton VIC Australia
| | - Moshe Olshansky
- Department of Microbiology, Immunity Theme, Biomedicine Discovery Institute Monash University Clayton VIC Australia
| | | | - Carla M Roots
- John Curtin School of Medical Research Australian National University Canberra ACT Australia
| | - Yovina Sontani
- John Curtin School of Medical Research Australian National University Canberra ACT Australia
| | - E Bridie Day
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity University of Melbourne Parkville VIC Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity University of Melbourne Parkville VIC Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity University of Melbourne Parkville VIC Australia
| | - Christopher C Goodnow
- John Curtin School of Medical Research Australian National University Canberra ACT Australia
- Garvan Institute of Medical Research & Cellular Genomics Futures Institute University of New South Wales Darlinghurst NSW Australia
| | - Brendan E Russ
- Department of Microbiology, Immunity Theme, Biomedicine Discovery Institute Monash University Clayton VIC Australia
| | - Stephen R Daley
- John Curtin School of Medical Research Australian National University Canberra ACT Australia
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health Queensland University of Technology Brisbane QLD Australia
| | - Stephen J Turner
- Department of Microbiology, Immunity Theme, Biomedicine Discovery Institute Monash University Clayton VIC Australia
| |
Collapse
|
37
|
This S, Paidassi H. New perspectives on the regulation of germinal center reaction via αvβ8- mediated activation of TGFβ. Front Immunol 2022; 13:942468. [PMID: 36072589 PMCID: PMC9441935 DOI: 10.3389/fimmu.2022.942468] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor-β (TGFβ) is a long-known modulator of immune responses but has seemingly contradictory effects on B cells. Among cytokines, TGFβ has the particularity of being produced and secreted in a latent form and must be activated before it can bind to its receptor and induce signaling. While the concept of controlled delivery of TGFβ signaling via αvβ8 integrin-mediated activation has gained some interest in the field of mucosal immunity, the role of this molecular mechanism in regulating T-dependent B cell responses is just emerging. We review here the role of TGFβ and its activation, in particular by αvβ8 integrin, in the regulation of mucosal IgA responses and its demonstrated and putative involvement in regulating germinal center (GC) B cell responses. We examine both the direct effect of TGFβ on GC B cells and its ability to modulate the functions of helper cells, namely follicular T cells (Tfh and Tfr) and follicular dendritic cells. Synthetizing recently published works, we reconcile apparently conflicting data and propose an innovative and unified view on the regulation of the GC reaction by TGFβ, highlighting the role of its activation by αvβ8 integrin.
Collapse
Affiliation(s)
- Sébastien This
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de microbiologie, immunologie et infectiologie, Université de Montréal, Montréal, QC, Canada
| | - Helena Paidassi
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| |
Collapse
|
38
|
Lai HZ, Han JR, Fu X, Ren YF, Li ZH, You FM. Targeted Approaches to HER2-Low Breast Cancer: Current Practice and Future Directions. Cancers (Basel) 2022; 14:cancers14153774. [PMID: 35954438 PMCID: PMC9367369 DOI: 10.3390/cancers14153774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary HER2-low breast cancer (BC) accounts for more than half of breast cancer patients. Anti-HER2 therapy has been ineffective in HER2-low BC, for which palliative chemotherapy is the main treatment modality. The definitive efficacy of T-Dxd in HER2-low BC breaks previous treatment strategies, which will redefine HER2-low and thus reshape anti-HER2 therapy. This review summarizes detection technologies and novel agents for HER2-low BC, and explores their possible role in future clinics, to provide ideas for the diagnosis and treatment of HER2-low BC. Abstract HER2-low breast cancer (BC) has a poor prognosis, making the development of more suitable treatment an unmet clinical need. While chemotherapy is the main method of treatment for HER2-low BC, not all patients benefit from it. Antineoplastic therapy without chemotherapy has shown promise in clinical trials and is being explored further. As quantitative detection techniques become more advanced, they assist in better defining the expression level of HER2 and in guiding the development of targeted therapies, which include directly targeting HER2 receptors on the cell surface, targeting HER2-related intracellular signaling pathways and targeting the immune microenvironment. A new anti-HER2 antibody-drug conjugate called T-DM1 has been successfully tested and found to be highly effective in clinical trials. With this progress, it could eventually be transformed from a disease without a defined therapeutic target into a disease with a defined therapeutic molecular target. Furthermore, efforts are being made to compare the sequencing and combination of chemotherapy, endocrine therapy, and HER2-targeted therapy to improve prognosis to customize the subtype of HER2 low expression precision treatment regimens. In this review, we summarize the current and upcoming treatment strategies, to achieve accurate management of HER2-low BC.
Collapse
|
39
|
Papadogkonas G, Papamatheakis DA, Spilianakis C. 3D Genome Organization as an Epigenetic Determinant of Transcription Regulation in T Cells. Front Immunol 2022; 13:921375. [PMID: 35812421 PMCID: PMC9257000 DOI: 10.3389/fimmu.2022.921375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
In the heart of innate and adaptive immunity lies the proper spatiotemporal development of several immune cell lineages. Multiple studies have highlighted the necessity of epigenetic and transcriptional regulation in cell lineage specification. This mode of regulation is mediated by transcription factors and chromatin remodelers, controlling developmentally essential gene sets. The core of transcription and epigenetic regulation is formulated by different epigenetic modifications determining gene expression. Apart from “classic” epigenetic modifications, 3D chromatin architecture is also purported to exert fundamental roles in gene regulation. Chromatin conformation both facilitates cell-specific factor binding at specified regions and is in turn modified as such, acting synergistically. The interplay between global and tissue-specific protein factors dictates the epigenetic landscape of T and innate lymphoid cell (ILC) lineages. The expression of global genome organizers such as CTCF, YY1, and the cohesin complexes, closely cooperate with tissue-specific factors to exert cell type-specific gene regulation. Special AT-rich binding protein 1 (SATB1) is an important tissue-specific genome organizer and regulator controlling both long- and short-range chromatin interactions. Recent indications point to SATB1’s cooperation with the aforementioned factors, linking global to tissue-specific gene regulation. Changes in 3D genome organization are of vital importance for proper cell development and function, while disruption of this mechanism can lead to severe immuno-developmental defects. Newly emerging data have inextricably linked chromatin architecture deregulation to tissue-specific pathophysiological phenotypes. The combination of these findings may shed light on the mechanisms behind pathological conditions.
Collapse
Affiliation(s)
- George Papadogkonas
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Dionysios-Alexandros Papamatheakis
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Charalampos Spilianakis
- Department of Biology, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- *Correspondence: Charalampos Spilianakis,
| |
Collapse
|
40
|
Ozawa T, Fujii K, Sudo T, Doi Y, Nakai R, Shingai Y, Ueda T, Baba Y, Hosen N, Yokota T. Special AT-Rich Sequence-Binding Protein 1 Supports Survival and Maturation of Naive B Cells Stimulated by B Cell Receptors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1937-1946. [PMID: 35379742 DOI: 10.4049/jimmunol.2101097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/09/2022] [Indexed: 11/19/2022]
Abstract
Epigenetic mechanisms underpin the elaborate activities of essential transcription factors in lymphocyte development. Special AT-rich sequence-binding protein 1 (SATB1) is a chromatin remodeler that orchestrates the spatial and temporal actions of transcription factors. Previous studies have revealed the significance of SATB1 in T cell lineage. However, whether and how SATB1 controls B cell lineage development is yet to be clarified. In this study, we show that SATB1 is an important factor during splenic B cell maturation. By analyzing SATB1/Tomato reporter mice, we determined the dynamic fluctuation of SATB1 expression in the B cell lineage. Although SATB1 expression decreased to minimal levels during B cell differentiation in the bone marrow, it resurged markedly in naive B cells in the spleen. The expression was dramatically downregulated upon Ag-induced activation. Splenic naive B cells were subdivided into two categories, namely SATB1high and SATB1-/low, according to their SATB1 expression levels. SATB1high naive B cells were less susceptible to death and greater proliferative than were SATB1-/low cells during incubation with an anti-IgM Ab. Additionally, SATB1high cells tended to induce the expression of MHC class II, CD86, and CD83. Accordingly, naive B cells from B lineage-specific SATB1 conditional knockout mice were more susceptible to apoptosis than that in the control group upon anti-IgM Ab stimulation in vitro. Furthermore, conditional knockout mice were less capable of producing Ag-specific B cells after immunization. Collectively, our findings suggest that SATB1 expression increases in naive B cells and plays an important role in their survival and maturation.
Collapse
Affiliation(s)
- Takayuki Ozawa
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kentaro Fujii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takao Sudo
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yukiko Doi
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ritsuko Nakai
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Shingai
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoaki Ueda
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Naoki Hosen
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan.,Laboratory of Cellular Immunotherapy, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan; and.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Takafumi Yokota
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan;
| |
Collapse
|
41
|
Kong Y, Jing Y, Allard D, Scavuzzo MA, Sprouse ML, Borowiak M, Bettini ML, Bettini M. A dormant T cell population with autoimmune potential exhibits low self-reactivity and infiltrates islets in type 1 diabetes. Eur J Immunol 2022; 52:1158-1170. [PMID: 35389516 DOI: 10.1002/eji.202149690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 11/09/2022]
Abstract
The contribution of low affinity T cells to autoimmunity in the context of polyclonal T cell responses is understudied due to the limitations in their capture by tetrameric reagents and low level of activation in response to antigenic stimulation. As a result, low affinity T cells are often disregarded as non-antigen specific cells irrelevant to the immune response. Our study aimed to assess how the level of self-antigen reactivity shapes T cell lineage and effector responses in the context of spontaneous tissue specific autoimmunity observed in NOD mice. Using multi-color flow cytometry in combination with Nur77GFP reporter of TCR signaling we identified a dormant population of T cells that infiltrated the pancreatic islets of pre-diabetic NOD mice, which exhibited reduced level of self-tissue reactivity based on expression of CD5 and Nur77GFP . We showed that these CD5low T cells had a unique TCR repertoire, exhibited low activation and minimal effector function; however, induced rapid diabetes upon transfer. The CD4+ CD5low T cell population displayed transcriptional signature of central memory T cells, consistent with the ability to acquire effector function post-transfer. Transcriptional profile of CD5low T cells was similar to T cells expressing a low affinity TCR, indicating TCR affinity to be the important factor in shaping CD5low T cell phenotype and function at the tissue site. Overall, our study suggests that autoimmune tissue can maintain a reservoir of undifferentiated central memory-like autoreactive T cells with pathogenic effector potential that might be an important source for effector T cells during long-term chronic autoimmunity. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yuelin Kong
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030
| | - Yi Jing
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112
| | - Denise Allard
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112
| | - Marissa A Scavuzzo
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Maran L Sprouse
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030
| | - Malgorzata Borowiak
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,McNair Medical Institute, Houston, TX, 77030
| | - Matthew L Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112.,McNair Medical Institute, Houston, TX, 77030
| | - Maria Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112.,McNair Medical Institute, Houston, TX, 77030
| |
Collapse
|
42
|
Schmidleithner L, Feuerer M. Tfh cells induce intratumoral tertiary lymphoid structures. Trends Immunol 2022; 43:274-276. [PMID: 35272934 DOI: 10.1016/j.it.2022.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 01/11/2023]
Abstract
Tertiary lymphoid structures (TLS) are highly organized ectopic structures found in nonlymphoid organs under chronic inflammatory conditions, including cancer. A recent study by Chaurio et al. reports that repression of Satb1 in CD4+ T cells can lead to increased Tfh cell differentiation, driving intratumoral TLS formation, resulting in reduced tumor growth in mice.
Collapse
Affiliation(s)
- Lisa Schmidleithner
- Division of Immunology, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Markus Feuerer
- Division of Immunology, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
43
|
Abstract
The cytokine, transforming growth factor beta (TGF-β), has a history of more than 40 years. TGF-β is secreted by many tumor cells and is associated with tumor growth and cancer immunity. The canonical TGF-β signaling pathway, SMAD, controls both tumor metastasis and immune regulation, thereby regulating cancer immunity. TGF-β regulates multiple types of immune cells in tumor microenvironment, including T cells, natural killer (NK) cells, and macrophages. One of the main roles of TGF-β in the tumor microenvironment is the generation of regulatory T cells, which contribute to the suppression of anti-tumor immunity. Because cancer is one of the highest causes of death globally, the discovery of immune checkpoint inhibitors by Honjo and Allison in cancer immunotherapy earned a Nobel Prize in 2018. TGF-β also regulates the levels of immune checkpoints inhibitory receptors on immune cells. Immune checkpoints inhibitors are now being developed along with anti-TGF-β antibody and/or TGF-β inhibitors. More recently, chimeric antigen receptors (CARs) were applied to cancer immunity and tried to combine with TGF-β blockers.
Collapse
Affiliation(s)
| | - WanJun Chen
- Mucosal Immunology Section, NIDCR, National Institute of Health
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Akita University Graduate School of Medicine
| |
Collapse
|
44
|
Xiao Y, Yang K, Wang Z, Zhao M, Deng Y, Ji W, Zou Y, Qian C, Liu Y, Xiao H, Liu H. CD44-Mediated Poor Prognosis in Glioma Is Associated With M2-Polarization of Tumor-Associated Macrophages and Immunosuppression. Front Surg 2022; 8:775194. [PMID: 35187044 PMCID: PMC8850306 DOI: 10.3389/fsurg.2021.775194] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/29/2021] [Indexed: 12/31/2022] Open
Abstract
Background Glioma is the most common primary brain tumor with a poor prognosis. Key genes that are negatively related to prognosis may provide the therapy targets to cure glioma. To clarify the role of CD44 in glioma, we explored its function at bulk-transcriptome, spatial and single-cell transcriptome levels. Methods In total, expression profiles with survival data of whole-grade glioma from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), RNA-seq data with anatomic information of glioblastoma (GBM) from the Ivy Glioblastoma Atlas Project, RNA-sequencing (RNA-seq) data from recurrent GBM receiving adjuvant anti-PD-1 immunotherapy accessed through GSE121810, and single-cell RNA-seq data of GBM under accession GSE103224 were enrolled in this study. CD44-specific findings were further analyzed by R language. Results CD44 is positively correlated with WHO grade of malignancy and is negatively related to prognosis in glioma. Meanwhile, CD44 predominantly expresses in GBM mesenchymal subtype, and gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses reveal that CD44 positively coexpressed genes are closely related to glioma immunity. Moreover, CD44+ cells mainly distribute in perinecrotic region with high expression of immune factors. At single-cell resolution, only malignant tumor cells, tumor-associated macrophages (TAMs), and T cells express CD44 in GBM. CD44+ malignant tumor cells are in mesenchymal-1-like (MES1-like) cellular state, and CD44+ TAMs are in M2 phenotype. CD44+ T cells have high expression of both PD-1 and PD-L1. CD44 and its directly interacted inhibitory immunomodulators are upregulated in patients with nonresponder recurrent GBM treated with PD-1 blockade therapy. Conclusion Our work demonstrates that CD44, a new M2 TAM biomarker, is involved in immune suppressor and promote glioma progression in glioma microenvironment. These results expand our understanding of CD44-specific clinical and immune features in glioma.
Collapse
Affiliation(s)
- Yong Xiao
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Kun Yang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zhen Wang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yanxiang Deng
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Wei Ji
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yuanjie Zou
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chunfa Qian
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yong Liu
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- *Correspondence: Hong Xiao
| | - Hongyi Liu
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Hongyi Liu
| |
Collapse
|
45
|
Chaurio RA, Anadon CM, Costich TL, Payne KK, Biswas S, Harro CM, Moran C, Ortiz AC, Cortina C, Rigolizzo KE, Sprenger KB, Mine JA, Innamarato PP, Mandal G, Powers JJ, Martin A, Wang Z, Mehta S, Perez BA, Li R, Robinson J, Kroeger JL, Curiel TJ, Yu X, Rodriguez PC, Conejo-Garcia JR. TGF-β-mediated silencing of genomic organizer SATB1 promotes Tfh cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity 2022; 55:115-128.e9. [PMID: 35021053 PMCID: PMC8852221 DOI: 10.1016/j.immuni.2021.12.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/17/2021] [Accepted: 12/08/2021] [Indexed: 01/13/2023]
Abstract
The immune checkpoint receptor PD-1 on T follicular helper (Tfh) cells promotes Tfh:B cell interactions and appropriate positioning within tissues. Here, we examined the impact of regulation of PD-1 expression by the genomic organizer SATB1 on Tfh cell differentiation. Vaccination of CD4CreSatb1f/f mice enriched for antigen-specific Tfh cells, and TGF-β-mediated repression of SATB1 enhanced Tfh differentiation of human T cells. Mechanistically, high Icos expression in Satb1-/- CD4+ T cells promoted Tfh cell differentiation by preventing T follicular regulatory cell skewing and resulted in increased isotype-switched B cell responses in vivo. Ovarian tumors in CD4CreSatb1f/f mice accumulated tumor antigen-specific, LIGHT+CXCL13+IL-21+ Tfh cells and tertiary lymphoid structures (TLS). TLS formation decreased tumor growth in a CD4+ T cell and CXCL13-dependent manner. The transfer of Tfh cells, but not naive CD4+ T cells, induced TLS at tumor beds and decreased tumor growth. Thus, TGF-β-mediated silencing of Satb1 licenses Tfh cell differentiation, providing insight into the genesis of TLS within tumors.
Collapse
Affiliation(s)
- Ricardo A Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carmen M Anadon
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Tara Lee Costich
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kyle K Payne
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carly M Harro
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carlos Moran
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Antonio C Ortiz
- Department of Analytic Microscopy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carla Cortina
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kristen E Rigolizzo
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kimberly B Sprenger
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jessica A Mine
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Pasquale P Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Gunjan Mandal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John J Powers
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Alexandra Martin
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Zhitao Wang
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sumit Mehta
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bradford A. Perez
- Department of Radiation Therapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John Robinson
- Department of Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jodi L Kroeger
- Department of Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Tyler J Curiel
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.,Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.,CORRESPONDENCE: Jose R Conejo-Garcia, MD, PhD (LEAD CONTACT), H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, , Phone: (813) 745-8282, Fax: (813) 745-5580
| |
Collapse
|
46
|
Li Z, Sun G, Sun G, Cheng Y, Wu L, Wang Q, Lv C, Zhou Y, Xia Y, Tang W. Various Uses of PD1/PD-L1 Inhibitor in Oncology: Opportunities and Challenges. Front Oncol 2021; 11:771335. [PMID: 34869005 PMCID: PMC8635629 DOI: 10.3389/fonc.2021.771335] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
The occurrence and development of cancer are closely related to the immune escape of tumor cells and immune tolerance. Unlike previous surgical, chemotherapy, radiotherapy and targeted therapy, tumor immunotherapy is a therapeutic strategy that uses various means to stimulate and enhance the immune function of the body, and ultimately achieves the goal of controlling tumor cells.With the in-depth understanding of tumor immune escape mechanism and tumor microenvironment, and the in-depth study of tumor immunotherapy, immune checkpoint inhibitors represented by Programmed Death 1/Programmed cell Death-Ligand 1(PD-1/PD-L1) inhibitors are becoming increasingly significant in cancer medication treatment. employ a variety of ways to avoid detection by the immune system, a single strategy is not more effective in overcoming tumor immune evasion and metastasis. Combining different immune agents or other drugs can effectively address situations where immunotherapy is not efficacious, thereby increasing the chances of success and alternative access to alternative immunotherapy. Immune combination therapies for cancer have become a hot topic in cancer treatment today. In this paper, several combination therapeutic modalities of PD1/PD-L1 inhibitors are systematically reviewed. Finally, an analysis and outlook are provided in the context of the recent advances in combination therapy with PD1/PD-L1 inhibitors and the pressing issues in this field.
Collapse
Affiliation(s)
- Zhitao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guoqiang Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guangshun Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ye Cheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liangliang Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yichan Zhou
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, China
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, China
| |
Collapse
|
47
|
Durślewicz J, Klimaszewska-Wiśniewska A, Jóźwicki J, Antosik P, Smolińska-Świtała M, Gagat M, Kowalewski A, Grzanka D. Prognostic Significance of TLR2, SMAD3 and Localization-dependent SATB1 in Stage I and II Non-Small-Cell Lung Cancer Patients. Cancer Control 2021; 28:10732748211056697. [PMID: 34818944 PMCID: PMC8640983 DOI: 10.1177/10732748211056697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study aimed to explore the prognostic value of SATB1, SMAD3, and TLR2 expression in non-small-cell lung carcinoma patients with clinical stages I-II. To investigate, we evaluated immunohistochemical staining to each of these markers using tissue sections from 69 patients from our cohort and gene expression data for The Cancer Genome Atlas (TCGA) cohort. We found that, in our cohort, high expression levels of nuclear SATB1n and SMAD3 were independent prognostic markers for better overall survival (OS) in NSCLC patients. Interestingly, expression of cytoplasmic SATB1c exhibited a significant but inverse association with survival rate, and it was an independent predictor of unfavorable prognosis. Likewise, TLR2 was a negative outcome biomarker for NSCLC even when adjusting for covariates. Importantly, stratification of NSCLCs with respect to combined expression of the three biomarkers allowed us to identify subgroups of patients with the greatest difference in duration of survival. Specifically, expression profile of SATB1n-high/SMAD3high/TLR2low was associated with the best OS, and it was superior to each single protein alone in predicting patient prognosis. Furthermore, based on the TCGA dataset, we found that overexpression of SATB1 mRNA was significantly associated with better OS, whereas high mRNA levels of SMAD3 and TLR2 with poor OS. In conclusion, the present study identified a set of proteins that may play a significant role in predicting prognosis of NSCLC patients with clinical stages I-II.
Collapse
Affiliation(s)
- Justyna Durślewicz
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| | - Anna Klimaszewska-Wiśniewska
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| | - Jakub Jóźwicki
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| | - Paulina Antosik
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| | - Marta Smolińska-Świtała
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| | - Maciej Gagat
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| | - Adam Kowalewski
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland.,Department of Tumor Pathology and Pathomorphology, Oncology Centre, Prof Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, 49577Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
48
|
Wang G, Tajima M, Honjo T, Ohta A. STAT5 interferes with PD-1 transcriptional activation and affects CD8+ T-cell sensitivity to PD-1-dependent immunoregulation. Int Immunol 2021; 33:563-572. [PMID: 34453440 DOI: 10.1093/intimm/dxab059] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/27/2021] [Indexed: 01/22/2023] Open
Abstract
Programmed cell death-1 (PD-1) is a co-inhibitory receptor that dampens immune responses upon interaction with PD-L1 and PD-L2. Although PD-1 expression on T cells is known to be activation-dependent, how cytokines modify its regulation is not fully resolved. Using polyclonal T-cell activation to study cytokine-dependent PD-1 regulation, we found that IL-2 inhibited transcriptional up-regulation of PD-1 despite the promotion of T-cell activation. The IL-2-mediated reduction in PD-1 expression augmented CD8+ T-cell activities against PD-L1-expressing target cells. To study the mechanism of PD-1 reduction, we focused on STAT5 activation in the IL-2 signaling pathway. Bioinformatic analysis suggested a novel conserved PD-1 promoter domain where NFAT and STAT5 can potentially compete with each other for binding. NFAT1 interaction with this domain revealed substantial potency in PD-1 transcription compared to STAT5A, and STAT5A overexpression could quench NFAT1-dependent PD-1 up-regulation in a sequence-specific manner. Chromatin immunoprecipitation analysis of activated T cells showed that IL-2 treatment significantly diminished the binding of NFAT1 and NFAT2 in the hypothesized competition site, while STAT5 binding to the same region was increased. These results raise the possibility that the competition of transcriptional factors might be involved in the fine-tuning of PD-1 expression by cytokines such as IL-2.
Collapse
Affiliation(s)
- Guanning Wang
- Department of Immunology, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan
| | - Masaki Tajima
- Department of Immunology, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Akio Ohta
- Department of Immunology, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan
| |
Collapse
|
49
|
Akbari B, Ghahri-Saremi N, Soltantoyeh T, Hadjati J, Ghassemi S, Mirzaei HR. Epigenetic strategies to boost CAR T cell therapy. Mol Ther 2021; 29:2640-2659. [PMID: 34365035 DOI: 10.1016/j.ymthe.2021.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/19/2021] [Accepted: 07/31/2021] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has led to a paradigm shift in cancer immunotherapy, but still several obstacles limit CAR T cell efficacy in cancers. Advances in high-throughput technologies revealed new insights into the role that epigenetic reprogramming plays in T cells. Mechanistic studies as well as comprehensive epigenome maps revealed an important role for epigenetic remodeling in T cell differentiation. These modifications shape the overall immune response through alterations in T cell phenotype and function. Here, we outline how epigenetic modifications in CAR T cells can overcome barriers limiting CAR T cell effectiveness, particularly in immunosuppressive tumor microenvironments. We also offer our perspective on how selected epigenetic modifications can boost CAR T cells to ultimately improve the efficacy of CAR T cell therapy.
Collapse
Affiliation(s)
- Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Navid Ghahri-Saremi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Tahereh Soltantoyeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran.
| |
Collapse
|
50
|
Renaude E, Kroemer M, Borg C, Peixoto P, Hervouet E, Loyon R, Adotévi O. Epigenetic Reprogramming of CD4 + Helper T Cells as a Strategy to Improve Anticancer Immunotherapy. Front Immunol 2021; 12:669992. [PMID: 34262562 PMCID: PMC8273698 DOI: 10.3389/fimmu.2021.669992] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/15/2021] [Indexed: 01/22/2023] Open
Abstract
Evidences highlight the role of various CD4+ helper T cells (CD4+ Th) subpopulations in orchestrating the immune responses against cancers. Epigenetics takes an important part in the regulation of CD4+ Th polarization and plasticity. In this review, we described the epigenetic factors that govern CD4+ T cells differentiation and recruitment in the tumor microenvironment and their subsequent involvement in the antitumor immunity. Finally, we discussed how to manipulate tumor reactive CD4+ Th responses by epigenetic drugs to improve anticancer immunotherapy.
Collapse
Affiliation(s)
- Elodie Renaude
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,Centre Hospitalier Universitaire de Besançon, Centre d'Investigation Clinique, INSERM CIC 1431, Besançon, France
| | - Marie Kroemer
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | - Christophe Borg
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,Centre Hospitalier Universitaire de Besançon, Centre d'Investigation Clinique, INSERM CIC 1431, Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Paul Peixoto
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,EPIGENEXP Platform, University of Bourgogne Franche-Comté, Besançon, France
| | - Eric Hervouet
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,EPIGENEXP Platform, University of Bourgogne Franche-Comté, Besançon, France.,DImaCell Platform, University of Bourgogne Franche-Comté, Besançon, France
| | - Romain Loyon
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Olivier Adotévi
- University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,Centre Hospitalier Universitaire de Besançon, Centre d'Investigation Clinique, INSERM CIC 1431, Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|